78 results on '"Srivatsan Raghavan"'
Search Results
2. Secondary IDH1 resistance mutations and oncogenic IDH2 mutations cause acquired resistance to ivosidenib in cholangiocarcinoma
- Author
-
James M. Cleary, Betty Rouaisnel, Antoine Daina, Srivatsan Raghavan, Lauren A. Roller, Brandon M. Huffman, Harshabad Singh, Patrick Y. Wen, Nabeel Bardeesy, Vincent Zoete, Brian M. Wolpin, and Julie-Aurore Losman
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract The mutant IDH1 inhibitor ivosidenib improves outcomes for patients with IDH1-mutated cholangiocarcinoma, but resistance inevitably develops. Mechanisms of resistance and strategies to overcome resistance are poorly understood. Here we describe two patients with IDH1 R132C-mutated metastatic cholangiocarcinoma who developed acquired resistance to ivosidenib. After disease progression, one patient developed an oncogenic IDH2 mutation, and the second patient acquired a secondary IDH1 D279N mutation. To characterize the putative IDH1 resistance mutation, cells expressing the double-mutant were generated. In vitro, IDH1 R132H/D279N produces (R)-2HG less efficiently than IDH1 R132H. However, its binding to ivosidenib is impaired and it retains the ability to produce (R)-2HG and promote cellular transformation in the presence of ivosidenib. The irreversible mutant IDH1 inhibitor LY3410738 binds and blocks (R)-2HG production and cellular transformation by IDH1 R132H/D279N. These resistance mechanisms suggest that IDH1-mutated cholangiocarcinomas remain dependent on (R)-2HG even after prolonged ivosidenib treatment. Sequential mutant IDH inhibitor therapy should be explored as a strategy to overcome acquired resistance to mutant IDH inhibitors.
- Published
- 2022
- Full Text
- View/download PDF
3. RNF43 G659fs is an oncogenic colorectal cancer mutation and sensitizes tumor cells to PI3K/mTOR inhibition
- Author
-
Lishan Fang, Dane Ford-Roshon, Max Russo, Casey O’Brien, Xiaozhe Xiong, Carino Gurjao, Maximilien Grandclaudon, Srivatsan Raghavan, Steven M. Corsello, Steven A. Carr, Namrata D. Udeshi, James Berstler, Ewa Sicinska, Kimmie Ng, and Marios Giannakis
- Subjects
Science - Abstract
The RNF43 G659fs mutation occurs frequently in colorectal cancer, but its function remains poorly understood. In this study, the authors show that RNF43 G659fs is an oncogenic colorectal cancer mutation and sensitizes tumor cells to PI3K/mTOR inhibition.
- Published
- 2022
- Full Text
- View/download PDF
4. Synthetic Lethal Interaction of SHOC2 Depletion with MEK Inhibition in RAS-Driven Cancers
- Author
-
Rita Sulahian, Jason J. Kwon, Katherine H. Walsh, Emma Pailler, Timothy L. Bosse, Maneesha Thaker, Diego Almanza, Joshua M. Dempster, Joshua Pan, Federica Piccioni, Nancy Dumont, Alfredo Gonzalez, Jonathan Rennhack, Behnam Nabet, John A. Bachman, Amy Goodale, Yenarae Lee, Mukta Bagul, Rosy Liao, Adrija Navarro, Tina L. Yuan, Raymond W.S. Ng, Srivatsan Raghavan, Nathanael S. Gray, Aviad Tsherniak, Francisca Vazquez, David E. Root, Ari J. Firestone, Jeff Settleman, William C. Hahn, and Andrew J. Aguirre
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: The mitogen-activated protein kinase (MAPK) pathway is a critical effector of oncogenic RAS signaling, and MAPK pathway inhibition may be an effective combination treatment strategy. We performed genome-scale loss-of-function CRISPR-Cas9 screens in the presence of a MEK1/2 inhibitor (MEKi) in KRAS-mutant pancreatic and lung cancer cell lines and identified genes that cooperate with MEK inhibition. While we observed heterogeneity in genetic modifiers of MEKi sensitivity across cell lines, several recurrent classes of synthetic lethal vulnerabilities emerged at the pathway level. Multiple members of receptor tyrosine kinase (RTK)-RAS-MAPK pathways scored as sensitizers to MEKi. In particular, we demonstrate that knockout, suppression, or degradation of SHOC2, a positive regulator of MAPK signaling, specifically cooperated with MEK inhibition to impair proliferation in RAS-driven cancer cells. The depletion of SHOC2 disrupted survival pathways triggered by feedback RTK signaling in response to MEK inhibition. Thus, these findings nominate SHOC2 as a potential target for combination therapy. : Sulahian, Kwon, and Walsh et al. performed several loss-of-function CRISPR-Cas9 screens in KRAS-mutant cancer cells treated with a MEK inhibitor and define the landscape of modifiers of MEK inhibitor sensitivity while highlighting that SHOC2 is a potent synthetic lethal target that serves as a critical signaling node to mediate MAP kinase pathway reactivation upon MEK inhibition. Keywords: Ras, KRAS, MEK inhibitor, synthetic lethal, SHOC2, CRISPR-Cas9 screen
- Published
- 2019
- Full Text
- View/download PDF
5. Correction: KEAP1 loss modulates sensitivity to kinase targeted therapy in lung cancer
- Author
-
Elsa Beyer Krall, Belinda Wang, Diana M Munoz, Nina Ilic, Srivatsan Raghavan, Matthew J Niederst, Kristine Yu, David A Ruddy, Andrew J Aguirre, Jong Wook Kim, Amanda J Redig, Justin F Gainor, Juliet A Williams, John M Asara, John G Doench, Pasi A Janne, Alice T Shaw, Robert E McDonald III III, Jeffrey A Engelman, Frank Stegmeier, Michael R Schlabach, and William C Hahn
- Subjects
Medicine ,Science ,Biology (General) ,QH301-705.5 - Published
- 2017
- Full Text
- View/download PDF
6. KEAP1 loss modulates sensitivity to kinase targeted therapy in lung cancer
- Author
-
Elsa B Krall, Belinda Wang, Diana M Munoz, Nina Ilic, Srivatsan Raghavan, Matthew J Niederst, Kristine Yu, David A Ruddy, Andrew J Aguirre, Jong Wook Kim, Amanda J Redig, Justin F Gainor, Juliet A Williams, John M Asara, John G Doench, Pasi A Janne, Alice T Shaw, Robert E McDonald III, Jeffrey A Engelman, Frank Stegmeier, Michael R Schlabach, and William C Hahn
- Subjects
CRISPR-Cas9 ,drug resistance ,lung cancer ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Inhibitors that target the receptor tyrosine kinase (RTK)/Ras/mitogen-activated protein kinase (MAPK) pathway have led to clinical responses in lung and other cancers, but some patients fail to respond and in those that do resistance inevitably occurs (Balak et al., 2006; Kosaka et al., 2006; Rudin et al., 2013; Wagle et al., 2011). To understand intrinsic and acquired resistance to inhibition of MAPK signaling, we performed CRISPR-Cas9 gene deletion screens in the setting of BRAF, MEK, EGFR, and ALK inhibition. Loss of KEAP1, a negative regulator of NFE2L2/NRF2, modulated the response to BRAF, MEK, EGFR, and ALK inhibition in BRAF-, NRAS-, KRAS-, EGFR-, and ALK-mutant lung cancer cells. Treatment with inhibitors targeting the RTK/MAPK pathway increased reactive oxygen species (ROS) in cells with intact KEAP1, and loss of KEAP1 abrogated this increase. In addition, loss of KEAP1 altered cell metabolism to allow cells to proliferate in the absence of MAPK signaling. These observations suggest that alterations in the KEAP1/NRF2 pathway may promote survival in the presence of multiple inhibitors targeting the RTK/Ras/MAPK pathway.
- Published
- 2017
- Full Text
- View/download PDF
7. Phase Ib and Expansion Study of Gemcitabine, Nab-Paclitaxel, and Ficlatuzumab in Patients With Metastatic Pancreatic Cancer
- Author
-
Kimberly Perez, Anna M Chiarella, James M Cleary, Nora Horick, Colin Weekes, Thomas Abrams, Lawrence Blaszkowsky, Peter Enzinger, Marios Giannakis, Lipika Goyal, Jeffrey A Meyerhardt, Douglas Rubinson, Matthew B Yurgelun, Wolfram Goessling, Bruce J Giantonio, Lauren Brais, Victoria Germon, Danielle Stonely, Srivatsan Raghavan, Basil Bakir, Koushik Das, Jason R Pitarresi, Andrew J Aguirre, Michael Needle, Anil K Rustgi, and Brian M Wolpin
- Subjects
Cancer Research ,Oncology - Abstract
Background In preclinical pancreatic ductal adenocarcinoma (PDAC) models, inhibition of hepatocyte growth factor (HGF) signaling using ficlatuzumab, a recombinant humanized anti-HGF antibody, and gemcitabine reduced tumor burden. Methods Patients with previously untreated metastatic PDAC enrolled in a phase Ib dose escalation study with 3 + 3 design of 2 dose cohorts of ficlatuzumab 10 and 20 mg/kg administered intravenously every other week with gemcitabine 1000 mg/m2 and albumin-bound paclitaxel 125 mg/m2 given 3 weeks on and 1 week off. This was followed by an expansion phase at the maximally tolerated dose of the combination. Results Twenty-six patients (sex, 12 male:14 female; median age, 68 years [range, 49-83 years]) were enrolled, 22 patients were evaluable. No dose–limiting toxicities were identified (N = 7 pts) and ficlatuzumab at 20 mg/kg was chosen as the maximum tolerated dose. Among the 21 patients treated at the MTD, best response by RECISTv1.1: 6 (29%) partial response, 12 (57%) stable disease, 1 (5%) progressive disease, and 2 (9%) not evaluable. Median progression-free survival and overall survival times were 11.0 months (95% CI, 7.6-11.4 months) and 16.2 months (95% CI, 9.1 months to not reached), respectively. Toxicities attributed to ficlatuzumab included hypoalbuminemia (grade 3, 16%; any grade, 52%) and edema (grade 3, 8%; any grade, 48%). Immunohistochemistry for c-Met pathway activation demonstrated higher tumor cell p-Met levels in patients who experienced response to therapy. Conclusion In this phase Ib trial, ficlatuzumab, gemcitabine, and albumin-bound paclitaxel were associated with durable treatment responses and increased rates of hypoalbuminemia and edema.
- Published
- 2023
8. MAF file for CNVs from Real-time Genomic Characterization of Advanced Pancreatic Cancer to Enable Precision Medicine
- Author
-
Brian M. Wolpin, Scott L. Carter, Ryan B. Corcoran, William C. Hahn, Jen Jen Yeh, Nikhil Wagle, David A. Tuveson, Paul B. Shyn, Jeffrey W. Miller, Robert J. Mayer, Matthew H. Kulke, Bruce E. Johnson, Jason L. Hornick, Gad Getz, Levi A. Garraway, Charles S. Fuchs, Matthew B. Yurgelun, Dean J. Welsch, Deborah Knoerzer, Richard B. Lanman, Rebecca J. Nagy, Stuart G. Silverman, Ewa Sicinska, Geoffrey I. Shapiro, Marvin Ryou, Douglas A. Rubinson, Michael H. Rosenthal, Kimberly Perez, Anuj K. Patel, Kimmie Ng, Janet E. Murphy, Jeffrey A. Meyerhardt, Nadine J. McCleary, Kunal Jajoo, Leona A. Doyle, James M. Cleary, Thomas Clancy, Kelly P. Burke, Joseph P. St. Pierre, Heather A. Shahzade, Emily E. Van Seventer, Brandon Nadres, Annacarolina Da Silva, Ana Babic, Nelly Oliver, Karla Helvie, Kristin Anderka, Lori Marini, Devin McCabe, Emma Reilly, Marisa W. Welch, Dorisanne Ragon, Lauren K. Brais, Jaegil Kim, Srivatsan Raghavan, Mehlika Hazar-Rethinam, Arezou A. Ghazani, Richard A. Moffitt, Nicholas D. Camarda, Jonathan A. Nowak, and Andrew J. Aguirre
- Abstract
MAF file for copy number variations identified in the cohort
- Published
- 2023
9. Supplemental Information and Figures S1-S7 from iRGD-guided Tumor-penetrating Nanocomplexes for Therapeutic siRNA Delivery to Pancreatic Cancer
- Author
-
Sangeeta N. Bhatia, Tyler Jacks, William C. Hahn, Charles S. Fuchs, Brian M. Wolpin, Andrew J. Aguirre, Felicia Hsu, Emilia M. Pulver, Ester J. Kwon, Srivatsan Raghavan, Mandar D. Muzumdar, Liangliang Hao, and Justin H. Lo
- Abstract
Figure S1. In vitro optimization of iRGD TPNs in PDAC cell lines; Figure S2. Dual knockdown using TPNs; Figure S3. In vitro and in vivo properties and function of PEGylated iRGD TPNs; Figure S4. Overview of organoid image analysis; Figure S5. iRGD TPN-mediated delivery of siRNA in models of pancreatic cancer; Figure S6. iRGD-mediated TPN uptake and NRP-1 expression in the tumor vasculature; Figure S7. Stromal elements in a KPC-derived orthotopic tumor section following iRGD TPN administration.
- Published
- 2023
10. Data from iRGD-guided Tumor-penetrating Nanocomplexes for Therapeutic siRNA Delivery to Pancreatic Cancer
- Author
-
Sangeeta N. Bhatia, Tyler Jacks, William C. Hahn, Charles S. Fuchs, Brian M. Wolpin, Andrew J. Aguirre, Felicia Hsu, Emilia M. Pulver, Ester J. Kwon, Srivatsan Raghavan, Mandar D. Muzumdar, Liangliang Hao, and Justin H. Lo
- Abstract
Pancreatic cancer is one of the leading causes of cancer-related death, with 5-year survival of 8.5%. The lack of significant progress in improving therapy reflects our inability to overcome the desmoplastic stromal barrier in pancreatic ductal adenocarcinoma (PDAC) as well as a paucity of new approaches targeting its genetic underpinnings. RNA interference holds promise in targeting key mutations driving PDAC; however, a nucleic acid delivery vehicle that homes to PDAC and breaches the stroma does not yet exist. Noting that the cyclic peptide iRGD mediates tumor targeting and penetration through interactions with αvβ3/5 integrins and neuropilin-1, we hypothesized that “tandem” peptides combining a cell-penetrating peptide and iRGD can encapsulate siRNA to form tumor-penetrating nanocomplexes (TPN) capable of delivering siRNA to PDAC. The use of directly conjugated iRGD is justified by receptor expression patterns in human PDAC biopsies. In this work, we optimize iRGD TPNs with polyethylene glycol (PEG)-peptide conjugates for systemic delivery to sites of disease. We show that TPNs effectively knockdown siRNA targets in PDAC cell lines and in an immunocompetent genetically engineered mouse model of PDAC. Furthermore, we validate their tumor-penetrating ability in three-dimensional organoids and autochthonous tumors. In murine therapeutic trials, TPNs delivering anti-Kras siRNA significantly delay tumor growth. Thus, iRGD TPNs hold promise in treating PDAC by not only overcoming physical barriers to therapy, but by leveraging the stroma to achieve knockdown of the gold-standard genetic target. Moreover, the modular construction of this delivery platform allows for facile adaptation to future genetic target candidates in pancreatic cancer. Mol Cancer Ther; 17(11); 2377–88. ©2018 AACR.
- Published
- 2023
11. Supplementary Table S4 from Real-time Genomic Characterization of Advanced Pancreatic Cancer to Enable Precision Medicine
- Author
-
Brian M. Wolpin, Scott L. Carter, Ryan B. Corcoran, William C. Hahn, Jen Jen Yeh, Nikhil Wagle, David A. Tuveson, Paul B. Shyn, Jeffrey W. Miller, Robert J. Mayer, Matthew H. Kulke, Bruce E. Johnson, Jason L. Hornick, Gad Getz, Levi A. Garraway, Charles S. Fuchs, Matthew B. Yurgelun, Dean J. Welsch, Deborah Knoerzer, Richard B. Lanman, Rebecca J. Nagy, Stuart G. Silverman, Ewa Sicinska, Geoffrey I. Shapiro, Marvin Ryou, Douglas A. Rubinson, Michael H. Rosenthal, Kimberly Perez, Anuj K. Patel, Kimmie Ng, Janet E. Murphy, Jeffrey A. Meyerhardt, Nadine J. McCleary, Kunal Jajoo, Leona A. Doyle, James M. Cleary, Thomas Clancy, Kelly P. Burke, Joseph P. St. Pierre, Heather A. Shahzade, Emily E. Van Seventer, Brandon Nadres, Annacarolina Da Silva, Ana Babic, Nelly Oliver, Karla Helvie, Kristin Anderka, Lori Marini, Devin McCabe, Emma Reilly, Marisa W. Welch, Dorisanne Ragon, Lauren K. Brais, Jaegil Kim, Srivatsan Raghavan, Mehlika Hazar-Rethinam, Arezou A. Ghazani, Richard A. Moffitt, Nicholas D. Camarda, Jonathan A. Nowak, and Andrew J. Aguirre
- Abstract
Treatment history for patients enrolled on the PancSeq protocol.
- Published
- 2023
12. Supplementary Data from FGFR2 Extracellular Domain In-Frame Deletions Are Therapeutically Targetable Genomic Alterations That Function as Oncogenic Drivers in Cholangiocarcinoma
- Author
-
Brian M. Wolpin, Nabeel Bardeesy, Vincent Zoete, Antoine Daina, Matthew Meyerson, Ryan B. Corcoran, Andrew D. Cherniack, William C. Hahn, Pasi A. Jänne, Deborah Schrag, Kimmie Ng, Claudio Zanna, Anna Pokorska-Bocci, Geoffrey I. Shapiro, Emma R. Hill, Sarah Denning, Rachel B. Keller, Ryan J. Sullivan, Anne Vaslin Chessex, Franck Brichory, Anuj K. Patel, Jiping Wang, Thomas E. Clancy, Thomas A. Abrams, Atul B. Shinagare, Maureen Loftus, Lauren K. Brais, Lei Shi, Lipika Goyal, Giulia Siravegna, Jonathan A. Nowak, Jason L. Hornick, Isobel J. Fetter, Douglas A. Rubinson, Hersh V. Gupta, Liam F. Spurr, Yvonne Y. Li, Qibiao Wu, Srivatsan Raghavan, and James M. Cleary
- Abstract
Supplemental Tables and Figures
- Published
- 2023
13. Supplementary Figures from Real-time Genomic Characterization of Advanced Pancreatic Cancer to Enable Precision Medicine
- Author
-
Brian M. Wolpin, Scott L. Carter, Ryan B. Corcoran, William C. Hahn, Jen Jen Yeh, Nikhil Wagle, David A. Tuveson, Paul B. Shyn, Jeffrey W. Miller, Robert J. Mayer, Matthew H. Kulke, Bruce E. Johnson, Jason L. Hornick, Gad Getz, Levi A. Garraway, Charles S. Fuchs, Matthew B. Yurgelun, Dean J. Welsch, Deborah Knoerzer, Richard B. Lanman, Rebecca J. Nagy, Stuart G. Silverman, Ewa Sicinska, Geoffrey I. Shapiro, Marvin Ryou, Douglas A. Rubinson, Michael H. Rosenthal, Kimberly Perez, Anuj K. Patel, Kimmie Ng, Janet E. Murphy, Jeffrey A. Meyerhardt, Nadine J. McCleary, Kunal Jajoo, Leona A. Doyle, James M. Cleary, Thomas Clancy, Kelly P. Burke, Joseph P. St. Pierre, Heather A. Shahzade, Emily E. Van Seventer, Brandon Nadres, Annacarolina Da Silva, Ana Babic, Nelly Oliver, Karla Helvie, Kristin Anderka, Lori Marini, Devin McCabe, Emma Reilly, Marisa W. Welch, Dorisanne Ragon, Lauren K. Brais, Jaegil Kim, Srivatsan Raghavan, Mehlika Hazar-Rethinam, Arezou A. Ghazani, Richard A. Moffitt, Nicholas D. Camarda, Jonathan A. Nowak, and Andrew J. Aguirre
- Abstract
Supplementary Figure S1: Overview of workflow and data generation. Supplementary Figure S2: Analysis of neoplastic cellularity. Supplementary Figure S3: Recurrent copy number alterations. Supplementary Figure S4: Mutational signature analysis. Supplementary Figure S5: Analysis of PDAC gene expression signatures. Supplementary Figure S6: Clinically relevant alterations in the cohort.
- Published
- 2023
14. Table S1 from TAS-120 Overcomes Resistance to ATP-Competitive FGFR Inhibitors in Patients with FGFR2 Fusion–Positive Intrahepatic Cholangiocarcinoma
- Author
-
Nabeel Bardeesy, Ryan B. Corcoran, Andrew X. Zhu, Dejan Juric, Gad Getz, Hiroshi Hirai, David T. Ting, Cyril H. Benes, William C. Hahn, A. John Iafrate, Alberto Bardelli, Robin K. Kelley, Rona Yaeger, James J. Harding, Vikram Deshpande, Kenneth K. Tanabe, Cristina R. Ferrone, Janet E. Murphy, Emily E. Van Seventer, Islam Baiev, Isobel J. Fetter, Ipsita Dey-Guha, Heather A. Shahzade, Brandon Nadres, Sachie Otsuki, Takeshi Sagara, Stephanie Reyes, Ron Arellano, Raul N. Uppot, Supriya K. Saha, Krushna C. Patra, Phuong Vu, Raymond W.S. Ng, Giulia Siravegna, Liudmila Elagina, Ignaty Leschiner, Srivatsan Raghavan, Jochen K. Lennerz, Ferran Fece de la Cruz, Leah Y. Liu, Lei Shi, and Lipika Goyal
- Abstract
Primer sequences
- Published
- 2023
15. Supplementary Experimental Methods from Real-time Genomic Characterization of Advanced Pancreatic Cancer to Enable Precision Medicine
- Author
-
Brian M. Wolpin, Scott L. Carter, Ryan B. Corcoran, William C. Hahn, Jen Jen Yeh, Nikhil Wagle, David A. Tuveson, Paul B. Shyn, Jeffrey W. Miller, Robert J. Mayer, Matthew H. Kulke, Bruce E. Johnson, Jason L. Hornick, Gad Getz, Levi A. Garraway, Charles S. Fuchs, Matthew B. Yurgelun, Dean J. Welsch, Deborah Knoerzer, Richard B. Lanman, Rebecca J. Nagy, Stuart G. Silverman, Ewa Sicinska, Geoffrey I. Shapiro, Marvin Ryou, Douglas A. Rubinson, Michael H. Rosenthal, Kimberly Perez, Anuj K. Patel, Kimmie Ng, Janet E. Murphy, Jeffrey A. Meyerhardt, Nadine J. McCleary, Kunal Jajoo, Leona A. Doyle, James M. Cleary, Thomas Clancy, Kelly P. Burke, Joseph P. St. Pierre, Heather A. Shahzade, Emily E. Van Seventer, Brandon Nadres, Annacarolina Da Silva, Ana Babic, Nelly Oliver, Karla Helvie, Kristin Anderka, Lori Marini, Devin McCabe, Emma Reilly, Marisa W. Welch, Dorisanne Ragon, Lauren K. Brais, Jaegil Kim, Srivatsan Raghavan, Mehlika Hazar-Rethinam, Arezou A. Ghazani, Richard A. Moffitt, Nicholas D. Camarda, Jonathan A. Nowak, and Andrew J. Aguirre
- Abstract
Supplementary Experimental Methods
- Published
- 2023
16. MAF file for mutations from Real-time Genomic Characterization of Advanced Pancreatic Cancer to Enable Precision Medicine
- Author
-
Brian M. Wolpin, Scott L. Carter, Ryan B. Corcoran, William C. Hahn, Jen Jen Yeh, Nikhil Wagle, David A. Tuveson, Paul B. Shyn, Jeffrey W. Miller, Robert J. Mayer, Matthew H. Kulke, Bruce E. Johnson, Jason L. Hornick, Gad Getz, Levi A. Garraway, Charles S. Fuchs, Matthew B. Yurgelun, Dean J. Welsch, Deborah Knoerzer, Richard B. Lanman, Rebecca J. Nagy, Stuart G. Silverman, Ewa Sicinska, Geoffrey I. Shapiro, Marvin Ryou, Douglas A. Rubinson, Michael H. Rosenthal, Kimberly Perez, Anuj K. Patel, Kimmie Ng, Janet E. Murphy, Jeffrey A. Meyerhardt, Nadine J. McCleary, Kunal Jajoo, Leona A. Doyle, James M. Cleary, Thomas Clancy, Kelly P. Burke, Joseph P. St. Pierre, Heather A. Shahzade, Emily E. Van Seventer, Brandon Nadres, Annacarolina Da Silva, Ana Babic, Nelly Oliver, Karla Helvie, Kristin Anderka, Lori Marini, Devin McCabe, Emma Reilly, Marisa W. Welch, Dorisanne Ragon, Lauren K. Brais, Jaegil Kim, Srivatsan Raghavan, Mehlika Hazar-Rethinam, Arezou A. Ghazani, Richard A. Moffitt, Nicholas D. Camarda, Jonathan A. Nowak, and Andrew J. Aguirre
- Abstract
MAF file for mutations identified in the cohort
- Published
- 2023
17. Figure S1 and S2 from TAS-120 Overcomes Resistance to ATP-Competitive FGFR Inhibitors in Patients with FGFR2 Fusion–Positive Intrahepatic Cholangiocarcinoma
- Author
-
Nabeel Bardeesy, Ryan B. Corcoran, Andrew X. Zhu, Dejan Juric, Gad Getz, Hiroshi Hirai, David T. Ting, Cyril H. Benes, William C. Hahn, A. John Iafrate, Alberto Bardelli, Robin K. Kelley, Rona Yaeger, James J. Harding, Vikram Deshpande, Kenneth K. Tanabe, Cristina R. Ferrone, Janet E. Murphy, Emily E. Van Seventer, Islam Baiev, Isobel J. Fetter, Ipsita Dey-Guha, Heather A. Shahzade, Brandon Nadres, Sachie Otsuki, Takeshi Sagara, Stephanie Reyes, Ron Arellano, Raul N. Uppot, Supriya K. Saha, Krushna C. Patra, Phuong Vu, Raymond W.S. Ng, Giulia Siravegna, Liudmila Elagina, Ignaty Leschiner, Srivatsan Raghavan, Jochen K. Lennerz, Ferran Fece de la Cruz, Leah Y. Liu, Lei Shi, and Lipika Goyal
- Abstract
Supplementary Figures and Legends
- Published
- 2023
18. Supplemental Video 1. Intravital imaging of iRGD TPNs in a xenograft model of pancreatic cancer from iRGD-guided Tumor-penetrating Nanocomplexes for Therapeutic siRNA Delivery to Pancreatic Cancer
- Author
-
Sangeeta N. Bhatia, Tyler Jacks, William C. Hahn, Charles S. Fuchs, Brian M. Wolpin, Andrew J. Aguirre, Felicia Hsu, Emilia M. Pulver, Ester J. Kwon, Srivatsan Raghavan, Mandar D. Muzumdar, Liangliang Hao, and Justin H. Lo
- Abstract
Intravital imaging of a subcutaneous pancreatic cancer xenograft (MIA PaCa-2) showing distribution of PEGylated iRGD TPNs (green) in tumor cell compartments surrounding abnormal tumor blood vessels. Collagen visualized through second-harmonic generation is displayed in magenta. Images in this video were captured at a depth of 60 μm from the edge of the exposed tumor. Each frame of 0.25 sec represents 5 min of elapsed time, starting 3 minutes after injection of the particles. Please refer to Figure S5 for reference scale bar.
- Published
- 2023
19. Data from FGFR2 Extracellular Domain In-Frame Deletions Are Therapeutically Targetable Genomic Alterations That Function as Oncogenic Drivers in Cholangiocarcinoma
- Author
-
Brian M. Wolpin, Nabeel Bardeesy, Vincent Zoete, Antoine Daina, Matthew Meyerson, Ryan B. Corcoran, Andrew D. Cherniack, William C. Hahn, Pasi A. Jänne, Deborah Schrag, Kimmie Ng, Claudio Zanna, Anna Pokorska-Bocci, Geoffrey I. Shapiro, Emma R. Hill, Sarah Denning, Rachel B. Keller, Ryan J. Sullivan, Anne Vaslin Chessex, Franck Brichory, Anuj K. Patel, Jiping Wang, Thomas E. Clancy, Thomas A. Abrams, Atul B. Shinagare, Maureen Loftus, Lauren K. Brais, Lei Shi, Lipika Goyal, Giulia Siravegna, Jonathan A. Nowak, Jason L. Hornick, Isobel J. Fetter, Douglas A. Rubinson, Hersh V. Gupta, Liam F. Spurr, Yvonne Y. Li, Qibiao Wu, Srivatsan Raghavan, and James M. Cleary
- Abstract
We conducted next-generation DNA sequencing on 335 biliary tract cancers and characterized the genomic landscape by anatomic site within the biliary tree. In addition to frequent FGFR2 fusions among patients with intrahepatic cholangiocarcinoma (IHCC), we identified FGFR2 extracellular domain in-frame deletions (EID) in 5 of 178 (2.8%) patients with IHCC, including two patients with FGFR2 p.H167_N173del. Expression of this FGFR2 EID in NIH3T3 cells resulted in constitutive FGFR2 activation, oncogenic transformation, and sensitivity to FGFR inhibitors. Three patients with FGFR2 EIDs were treated with Debio 1347, an oral FGFR1/2/3 inhibitor, and all showed partial responses. One patient developed an acquired L618F FGFR2 kinase domain mutation at disease progression and experienced a further partial response for 17 months to an irreversible FGFR2 inhibitor, futibatinib. Together, these findings reveal FGFR2 EIDs as an alternative mechanism of FGFR2 activation in IHCC that predicts sensitivity to FGFR inhibitors in the clinic.Significance:FGFR2 EIDs are transforming genomic alterations that occur predominantly in patients with IHCC. These FGFR2 EIDs are sensitive to FGFR inhibition in vitro, and patients with these alterations benefited from treatment with FGFR inhibitors in the clinic.This article is highlighted in the In This Issue feature, p. 2355
- Published
- 2023
20. Data from TAS-120 Overcomes Resistance to ATP-Competitive FGFR Inhibitors in Patients with FGFR2 Fusion–Positive Intrahepatic Cholangiocarcinoma
- Author
-
Nabeel Bardeesy, Ryan B. Corcoran, Andrew X. Zhu, Dejan Juric, Gad Getz, Hiroshi Hirai, David T. Ting, Cyril H. Benes, William C. Hahn, A. John Iafrate, Alberto Bardelli, Robin K. Kelley, Rona Yaeger, James J. Harding, Vikram Deshpande, Kenneth K. Tanabe, Cristina R. Ferrone, Janet E. Murphy, Emily E. Van Seventer, Islam Baiev, Isobel J. Fetter, Ipsita Dey-Guha, Heather A. Shahzade, Brandon Nadres, Sachie Otsuki, Takeshi Sagara, Stephanie Reyes, Ron Arellano, Raul N. Uppot, Supriya K. Saha, Krushna C. Patra, Phuong Vu, Raymond W.S. Ng, Giulia Siravegna, Liudmila Elagina, Ignaty Leschiner, Srivatsan Raghavan, Jochen K. Lennerz, Ferran Fece de la Cruz, Leah Y. Liu, Lei Shi, and Lipika Goyal
- Abstract
ATP-competitive fibroblast growth factor receptor (FGFR) kinase inhibitors, including BGJ398 and Debio 1347, show antitumor activity in patients with intrahepatic cholangiocarcinoma (ICC) harboring activating FGFR2 gene fusions. Unfortunately, acquired resistance develops and is often associated with the emergence of secondary FGFR2 kinase domain mutations. Here, we report that the irreversible pan-FGFR inhibitor TAS-120 demonstrated efficacy in 4 patients with FGFR2 fusion–positive ICC who developed resistance to BGJ398 or Debio 1347. Examination of serial biopsies, circulating tumor DNA (ctDNA), and patient-derived ICC cells revealed that TAS-120 was active against multiple FGFR2 mutations conferring resistance to BGJ398 or Debio 1347. Functional assessment and modeling the clonal outgrowth of individual resistance mutations from polyclonal cell pools mirrored the resistance profiles observed clinically for each inhibitor. Our findings suggest that strategic sequencing of FGFR inhibitors, guided by serial biopsy and ctDNA analysis, may prolong the duration of benefit from FGFR inhibition in patients with FGFR2 fusion–positive ICC.Significance:ATP-competitive FGFR inhibitors (BGJ398, Debio 1347) show efficacy in FGFR2-altered ICC; however, acquired FGFR2 kinase domain mutations cause drug resistance and tumor progression. We demonstrate that the irreversible FGFR inhibitor TAS-120 provides clinical benefit in patients with resistance to BGJ398 or Debio 1347 and overcomes several FGFR2 mutations in ICC models.This article is highlighted in the In This Issue feature, p. 983
- Published
- 2023
21. Data from Spatially Resolved Single-Cell Assessment of Pancreatic Cancer Expression Subtypes Reveals Co-expressor Phenotypes and Extensive Intratumoral Heterogeneity
- Author
-
Brian M. Wolpin, Jonathan A. Nowak, Andrew J. Aguirre, Alex K. Shalek, William C. Hahn, Aram F. Hezel, Albert C. Koong, Daniel T. Chang, Richard F. Dunne, David C. Linehan, Margaret M. Kozak, Emma R. Hill, Lauren K. Brais, Joseph D. Mancias, Jiping Wang, Thomas E. Clancy, James M. Cleary, Kimberly Perez, Harshabad Singh, Douglas A. Rubinson, Vicente Morales-Oyarvide, Dalia Elganainy, Mai Chan Lau, Kristen E. Lowder, Radha L. Kalekar, Timothy L. Bosse, Annan Yang, Junning Wang, Andrew W. Navia, Chen Yuan, Juha P. Väyrynen, Sara A. Väyrynen, Scott P. Ginebaugh, Kevin S. Kapner, Peter S. Winter, Srivatsan Raghavan, Jinming Zhang, Andressa Dias Costa, and Hannah L. Williams
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) has been classified into classical and basal-like transcriptional subtypes by bulk RNA measurements. However, recent work has uncovered greater complexity to transcriptional subtypes than was initially appreciated using bulk RNA expression profiling. To provide a deeper understanding of PDAC subtypes, we developed a multiplex immunofluorescence (mIF) pipeline that quantifies protein expression of six PDAC subtype markers (CLDN18.2, TFF1, GATA6, KRT17, KRT5, and S100A2) and permits spatially resolved, single-cell interrogation of pancreatic tumors from resection specimens and core needle biopsies. Both primary and metastatic tumors displayed striking intratumoral subtype heterogeneity that was associated with patient outcomes, existed at the scale of individual glands, and was significantly reduced in patient-derived organoid cultures. Tumor cells co-expressing classical and basal markers were present in > 90% of tumors, existed on a basal-classical polarization continuum, and were enriched in tumors containing a greater admixture of basal and classical cell populations. Cell–cell neighbor analyses within tumor glands further suggested that co-expressor cells may represent an intermediate state between expression subtype poles. The extensive intratumoral heterogeneity identified through this clinically applicable mIF pipeline may inform prognosis and treatment selection for patients with PDAC.Significance:A high-throughput pipeline using multiplex immunofluorescence in pancreatic cancer reveals striking expression subtype intratumoral heterogeneity with implications for therapy selection and identifies co-expressor cells that may serve as intermediates during subtype switching.
- Published
- 2023
22. Supplementary Data from Spatially Resolved Single-Cell Assessment of Pancreatic Cancer Expression Subtypes Reveals Co-expressor Phenotypes and Extensive Intratumoral Heterogeneity
- Author
-
Brian M. Wolpin, Jonathan A. Nowak, Andrew J. Aguirre, Alex K. Shalek, William C. Hahn, Aram F. Hezel, Albert C. Koong, Daniel T. Chang, Richard F. Dunne, David C. Linehan, Margaret M. Kozak, Emma R. Hill, Lauren K. Brais, Joseph D. Mancias, Jiping Wang, Thomas E. Clancy, James M. Cleary, Kimberly Perez, Harshabad Singh, Douglas A. Rubinson, Vicente Morales-Oyarvide, Dalia Elganainy, Mai Chan Lau, Kristen E. Lowder, Radha L. Kalekar, Timothy L. Bosse, Annan Yang, Junning Wang, Andrew W. Navia, Chen Yuan, Juha P. Väyrynen, Sara A. Väyrynen, Scott P. Ginebaugh, Kevin S. Kapner, Peter S. Winter, Srivatsan Raghavan, Jinming Zhang, Andressa Dias Costa, and Hannah L. Williams
- Abstract
Supplementary Figures
- Published
- 2023
23. Compressed phenotypic screens for complex multicellular models and high-content assays
- Author
-
Benjamin E. Mead, Conner Kummerlowe, Nuo Liu, Walaa E. Kattan, Thomas Cheng, Jaime H. Cheah, Christian K. Soule, Josh Peters, Kristen E. Lowder, Paul C. Blainey, William C. Hahn, Brian Cleary, Bryan Bryson, Peter S. Winter, Srivatsan Raghavan, and Alex K. Shalek
- Subjects
Article - Abstract
High-throughput phenotypic screens leveraging biochemical perturbations, high-content readouts, and complex multicellular models could advance therapeutic discovery yet remain constrained by limitations of scale. To address this, we establish a method for compressing screens by pooling perturbations followed by computational deconvolution. Conducting controlled benchmarks with a highly bioactive small molecule library and a high-content imaging readout, we demonstrate increased efficiency for compressed experimental designs compared to conventional approaches. To prove generalizability, we apply compressed screening to examine transcriptional responses of patient-derived pancreatic cancer organoids to a library of tumor-microenvironment (TME)-nominated recombinant protein ligands. Using single-cell RNA-seq as a readout, we uncover reproducible phenotypic shifts induced by ligands that correlate with clinical features in larger datasets and are distinct from reference signatures available in public databases. In sum, our approach enables phenotypic screens that interrogate complex multicellular models with rich phenotypic readouts to advance translatable drug discovery as well as basic biology.
- Published
- 2023
24. Opportunities for Utilization of DNA Repair Inhibitors in Homologous Recombination Repair-Deficient and Proficient Pancreatic Adenocarcinoma
- Author
-
Alan D. D'Andrea, James M. Cleary, Nilay Sethi, Harshabad Singh, Brandon M. Huffman, Jonathan A. Nowak, Stephanie K. Dougan, Andrew J. Aguirre, Geoffrey I. Shapiro, Brian M. Wolpin, and Srivatsan Raghavan
- Subjects
Cancer Research ,DNA Repair ,DNA damage ,business.industry ,DNA repair ,Poly ADP ribose polymerase ,Recombinational DNA Repair ,Adenocarcinoma ,Poly(ADP-ribose) Polymerase Inhibitors ,medicine.disease ,Article ,Germline ,Pancreatic Neoplasms ,chemistry.chemical_compound ,Oncology ,chemistry ,Pancreatic cancer ,medicine ,Cancer research ,Humans ,Homologous Recombination ,business ,Homologous recombination ,DNA - Abstract
Pancreatic cancer is rapidly progressive and notoriously difficult to treat with cytotoxic chemotherapy and targeted agents. Recent demonstration of the efficacy of maintenance PARP inhibition in germline BRCA mutated pancreatic cancer has raised hopes that increased understanding of the DNA damage response pathway will lead to new therapies in both homologous recombination (HR) repair-deficient and proficient pancreatic cancer. Here, we review the potential mechanisms of exploiting HR deficiency, replicative stress, and DNA damage-mediated immune activation through targeted inhibition of DNA repair regulatory proteins.
- Published
- 2021
25. Visualization of tuberculosis patient and Mycobacterium tuberculosis complex genotype data via host-pathogen maps.
- Author
-
Kristin P. Bennett, Cagri Ozcaglar, Janani Ranganathan, Srivatsan Raghavan, Jacob Katz, Dan Croft, Bülent Yener, and Amina Shabbeer
- Published
- 2011
- Full Text
- View/download PDF
26. Spatially-resolved single-cell assessment of pancreatic cancer expression subtypes reveals co-expressor phenotypes and extensive intra-tumoral heterogeneity
- Author
-
Hannah L. Williams, Andressa Dias Costa, Jinming Zhang, Srivatsan Raghavan, Peter S. Winter, Kevin S. Kapner, Scott P. Ginebaugh, Sara A. Väyrynen, Juha P. Väyrynen, Chen Yuan, Andrew W. Navia, Junning Wang, Annan Yang, Timothy L. Bosse, Radha L. Kalekar, Kristen E. Lowder, Mai Chan Lau, Dalia Elganainy, Vicente Morales-Oyarvide, Douglas A. Rubinson, Harshabad Singh, Kimberly Perez, James M. Cleary, Thomas E. Clancy, Jiping Wang, Joseph D. Mancias, Lauren K. Brais, Emma R. Hill, Margaret M. Kozak, David C. Linehan, Richard F. Dunne, Daniel T. Chang, Albert C. Koong, Aram F. Hezel, William C. Hahn, Alex K. Shalek, Andrew J. Aguirre, Jonathan A. Nowak, and Brian M. Wolpin
- Subjects
Cancer Research ,Oncology - Abstract
Pancreatic ductal adenocarcinoma (PDAC) has been classified into classical and basal-like transcriptional subtypes by bulk RNA measurements. However, recent work has uncovered greater complexity to transcriptional subtypes than was initially appreciated using bulk RNA expression profiling. To provide a deeper understanding of PDAC subtypes, we developed a multiplex immunofluorescence (mIF) pipeline that quantifies protein expression of six PDAC subtype markers (CLDN18.2, TFF1, GATA6, KRT17, KRT5, and S100A2) and permits spatially resolved, single-cell interrogation of pancreatic tumors from resection specimens and core needle biopsies. Both primary and metastatic tumors displayed striking intratumoral subtype heterogeneity that was associated with patient outcomes, existed at the scale of individual glands, and was significantly reduced in patient-derived organoid cultures. Tumor cells co-expressing classical and basal markers were present in > 90% of tumors, existed on a basal-classical polarization continuum, and were enriched in tumors containing a greater admixture of basal and classical cell populations. Cell–cell neighbor analyses within tumor glands further suggested that co-expressor cells may represent an intermediate state between expression subtype poles. The extensive intratumoral heterogeneity identified through this clinically applicable mIF pipeline may inform prognosis and treatment selection for patients with PDAC. Significance: A high-throughput pipeline using multiplex immunofluorescence in pancreatic cancer reveals striking expression subtype intratumoral heterogeneity with implications for therapy selection and identifies co-expressor cells that may serve as intermediates during subtype switching.
- Published
- 2022
27. The efficiency of dehydrating desiccants by centrifugation: An assessment of superabsorbent polymers
- Author
-
Alexandra Pine, Cheng Chi Wu, Brian J. Love, and Srivatsan Raghavan
- Subjects
Desiccant ,Sodium Acrylate ,Chromatography ,Chemistry ,General Chemical Engineering ,medicine.disease ,Synthetic urine ,Superabsorbent polymer ,Desorption ,Copolymer ,medicine ,Centrifugation ,Dehydration ,Physical and Theoretical Chemistry - Abstract
The centrifugal drying response from a sodium acrylate superabsorbent copolymer saturated with water, saline, and a synthetic urine mimic was evaluated. It was hypothesized that drying using mechan...
- Published
- 2021
28. Decision letter: Gallbladder adenocarcinomas undergo subclonal diversification and selection from precancerous lesions to metastatic tumors
- Author
-
Srivatsan Raghavan
- Published
- 2022
29. FGFR2Extracellular Domain In-Frame Deletions Are Therapeutically Targetable Genomic Alterations That Function as Oncogenic Drivers in Cholangiocarcinoma
- Author
-
Giulia Siravegna, Thomas A. Abrams, Isobel J Fetter, Atul B. Shinagare, Kimmie Ng, Sarah L. Denning, William C. Hahn, Maureen Loftus, Anuj K. Patel, Yvonne Y. Li, James M. Cleary, Liam F. Spurr, Srivatsan Raghavan, Ryan J. Sullivan, Douglas A. Rubinson, Matthew Meyerson, Hersh Gupta, Nabeel Bardeesy, Emma R. Hill, Antoine Daina, Brian M. Wolpin, Ryan B. Corcoran, Jiping Wang, Lauren K. Brais, Pasi A. Jänne, Claudio Zanna, Qibiao Wu, Lipika Goyal, Andrew D. Cherniack, Lei Shi, Anne Vaslin Chessex, Jonathan A. Nowak, Anna Pokorska-Bocci, Rachel B. Keller, Thomas E. Clancy, Jason L. Hornick, Vincent Zoete, Geoffrey I. Shapiro, Deborah Schrag, and Franck Brichory
- Subjects
musculoskeletal diseases ,0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Mutation ,integumentary system ,business.industry ,medicine.disease_cause ,Article ,DNA sequencing ,stomatognathic diseases ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Protein kinase domain ,Biliary tract ,Fibroblast growth factor receptor ,030220 oncology & carcinogenesis ,embryonic structures ,Extracellular ,Cancer research ,medicine ,business ,Function (biology) ,Intrahepatic Cholangiocarcinoma - Abstract
We conducted next-generation DNA sequencing on 335 biliary tract cancers and characterized the genomic landscape by anatomic site within the biliary tree. In addition to frequent FGFR2 fusions among patients with intrahepatic cholangiocarcinoma (IHCC), we identified FGFR2 extracellular domain in-frame deletions (EID) in 5 of 178 (2.8%) patients with IHCC, including two patients with FGFR2 p.H167_N173del. Expression of this FGFR2 EID in NIH3T3 cells resulted in constitutive FGFR2 activation, oncogenic transformation, and sensitivity to FGFR inhibitors. Three patients with FGFR2 EIDs were treated with Debio 1347, an oral FGFR1/2/3 inhibitor, and all showed partial responses. One patient developed an acquired L618F FGFR2 kinase domain mutation at disease progression and experienced a further partial response for 17 months to an irreversible FGFR2 inhibitor, futibatinib. Together, these findings reveal FGFR2 EIDs as an alternative mechanism of FGFR2 activation in IHCC that predicts sensitivity to FGFR inhibitors in the clinic.Significance:FGFR2 EIDs are transforming genomic alterations that occur predominantly in patients with IHCC. These FGFR2 EIDs are sensitive to FGFR inhibition in vitro, and patients with these alterations benefited from treatment with FGFR inhibitors in the clinic.This article is highlighted in the In This Issue feature, p. 2355
- Published
- 2021
30. USP9X mediates an acute adaptive response to MAPK suppression in pancreatic cancer but creates multiple actionable therapeutic vulnerabilities
- Author
-
Naiara Perurena, Rebecca Lock, Rachel A. Davis, Srivatsan Raghavan, Natalie F. Pilla, Raymond Ng, Patrick Loi, Caroline J. Guild, Abigail L. Miller, Ewa Sicinska, James M. Cleary, Douglas A. Rubinson, Brian M. Wolpin, Nathanael S. Gray, Sandro Santagata, William C. Hahn, Jennifer P. Morton, Owen J. Sansom, Andrew J. Aguirre, and Karen Cichowski
- Subjects
General Biochemistry, Genetics and Molecular Biology - Published
- 2023
31. Improved multicast routing with delay and delay variation constraints.
- Author
-
Sanjiv Kapoor and Srivatsan Raghavan
- Published
- 2000
- Full Text
- View/download PDF
32. Discovery of a selective inhibitor of doublecortin like kinase 1
- Author
-
Jinhua Wang, Senthil Muthaswamy, Kevin M. Haigis, Rita Sulahian, Kenneth D. Westover, Emily J. Poulin, Ryoma Ohi, Sergio Espinosa, Shuning He, Taebo Sim, Lianbo Li, Miljan Kuljanin, Nathanael S. Gray, Yan Liu, Nam Doo Kim, Joseph D. Mancias, Ling Huang, Brian M. Wolpin, Charles Y. Lin, Andrew J. Aguirre, Srivatsan Raghavan, Nora Diéguez-Martínez, Radha L. Kalekar, Jost Vrabic Koren, Zeng Zhiyang, Fleur M. Ferguson, James D Vasta, Behnam Nabet, William C. Hahn, Raymond W.S. Ng, Cesear Corona, Wayne Harshbarger, Alan L. Leggett, Matthew B. Robers, A. Thomas Look, Jose M. Lizcano, and Annan Yang
- Subjects
Male ,Proteomics ,Doublecortin Protein ,Protein Serine-Threonine Kinases ,Biology ,Article ,Transcriptome ,Mice ,Structure-Activity Relationship ,03 medical and health sciences ,Doublecortin-Like Kinases ,Downregulation and upregulation ,Cell Movement ,Cell Line, Tumor ,Organoid ,Animals ,Humans ,Protein Kinase Inhibitors ,Molecular Biology ,Zebrafish ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,Molecular Structure ,Kinase ,030302 biochemistry & molecular biology ,Intracellular Signaling Peptides and Proteins ,Phosphoproteomics ,Cell Biology ,Rats ,Molecular Docking Simulation ,Pancreatic Neoplasms ,Gene Expression Regulation ,Protein kinase domain ,Cancer research ,Drug Screening Assays, Antitumor ,Carcinoma, Pancreatic Ductal - Abstract
Doublecortin like kinase 1 (DCLK1) is an understudied kinase that is upregulated in a wide range of cancers, including pancreatic ductal adenocarcinoma (PDAC). However, little is known about its potential as a therapeutic target. We used chemoproteomic profiling and structure-based design to develop a selective, in vivo-compatible chemical probe of the DCLK1 kinase domain, DCLK1-IN-1. We demonstrate activity of DCLK1-IN-1 against clinically relevant patient-derived PDAC organoid models and use a combination of RNA-sequencing, proteomics and phosphoproteomics analysis to reveal that DCLK1 inhibition modulates proteins and pathways associated with cell motility in this context. DCLK1-IN-1 will serve as a versatile tool to investigate DCLK1 biology and establish its role in cancer. A highly selective inhibitor of the DCLK1/2 kinases is used to uncover the consequences of DCLK1 inhibition on viability, phosphosignaling and the transcriptome in patient-derived organoid models of pancreatic ductal adenocarcinoma.
- Published
- 2020
33. Abstract PR009: RNF43 G659fs is an oncogenic and immune-modulating colorectal cancer mutation and sensitizes tumor cells to PI3K/mTOR inhibition
- Author
-
Marios Giannakis, Lishan Fang, Dane Ford-Roshon, Max Russo, Casey O'Brien, Xiaozhe Xiong, Carino Gurjao, Maximilien Grandclaudon, Srivatsan Raghavan, Steven M. Corsello, Steven A. Carr, Namrata Udeshi, James Berstler, Ewa Sicinska, and Kimmie Ng
- Subjects
Cancer Research ,Oncology - Abstract
Introduction: The RNF43 p.G659fs mutation occurs with a frequency of approximately 8% in colorectal cancer (CRC) and is enriched among microsatellite-instability high tumors. However, despite RNF43 being implicated as a Wnt-signaling tumor suppressor, the function of the RNF43 p.G659fs mutation remains undescribed, with no specific therapies directed against it. We hypothesized that large-scale drug screening coupled with functional validation and testing in CRC pre-clinical models would reveal the functional role and selective vulnerabilities of this alteration, leading to new therapeutic approaches in CRC. Methods: We targeted the RNF43 659 site with CRISPR-Cas9 to generate isogenic RNF43659mut clones of CRC cell lines. We leveraged the Broad Institute’s drug repurposing library of over 5,300 compounds to identify selective hits (z5% selective killing in mutant vs WT) in RNF43659mut isogenic cells. These hits were validated in secondary screens and top drugs were tested in patient-derived organoid and xenograft models of the RNF43 p.G659fs mutation. We assessed the functional role of the mutation using cell viability and colony formation assays, quantitative mass spectrometry proteomics, immunoprecipitation and ubiquitination experiments. The immunomodulatory role of RNF43659mut was assessed through bulk and single-cell RNA Seq analysis of mutant cell lines and patient-derived CRC specimens. Results: Using edited cell line models, we demonstrated that RNF43 p.G659fs has a Wnt-independent oncogenic role in CRC. We performed a comprehensive drug screen in isogenic RNF43659mut HT29 CRC cells with a repurposing library of 5,363 compounds and discovered that RNF43659mut is selectively targeted by PI3K/mTOR inhibitors (p = 1.34 x 10-5, chi-square test). Secondary screens in LS513 RNF43659mut-edited and HCT116 RNF43G659fs-overexpressing cells confirmed these results and we further showed that alpelisib and PF-04691502 (a dual PI3K/mTOR inhibitor) selectively killed patient-derived organoids with the RNF43 p.G659Vfs mutation and reduced tumor growth in RNF43659mut CDX mice (p=0.0005, Student’s t-test). RNF43 p.G659fs increased PI3K/AKT signaling in vitro and in vivo through binding, ubiquitinating and degrading p85 and this was reversed by PI3K/mTOR inhibition, RNF43 knockout or RSPO stimulation. Additionally, in pre-clinical models and single-cell studies of human CRCs (>350,000 single cells), we found that RNF43659mut modulates interferon activity and NK cells in the CRC tumor microenvironment. Conclusions: Through functional and pharmacologic studies in pre-clinical models and human CRCs, we described that RNF43 p.G659fs activates PI3K/AKT signaling and modulates the CRC immune microenvironment. We showed that these effects are reversed with PI3K/mTOR inhibitors and that this class of drugs selectively targets RNF43659mut CRCs. Our results have important therapeutic implications for patients with CRC. Citation Format: Marios Giannakis, Lishan Fang, Dane Ford-Roshon, Max Russo, Casey O'Brien, Xiaozhe Xiong, Carino Gurjao, Maximilien Grandclaudon, Srivatsan Raghavan, Steven M. Corsello, Steven A. Carr, Namrata Udeshi, James Berstler, Ewa Sicinska, Kimmie Ng. RNF43 G659fs is an oncogenic and immune-modulating colorectal cancer mutation and sensitizes tumor cells to PI3K/mTOR inhibition [abstract]. In: Proceedings of the AACR Special Conference on Colorectal Cancer; 2022 Oct 1-4; Portland, OR. Philadelphia (PA): AACR; Cancer Res 2022;82(23 Suppl_1):Abstract nr PR009.
- Published
- 2022
34. Abstract A001: Clinical-genomic analysis of KRAS wild-type pancreatic cancer confirms alternative targetable drivers and provides insight for age and risk related clinical stratification
- Author
-
Harshabad Singh, Rachel B. Keller, Kevin S. Kapner, Julien Dilly, Srivatsan Raghavan, Chen Yuan, Eizabeth Cohen, Michael Tolstorukov, Emma Hill, Elizabeth Andrews, Lauren K. Brais, Annacarolina Da Silva, Kimberly Perez, Douglas A. Rubinson, Benjamin L. Schlechter, Michael H. Rosenthal, Jason L. Hornick, Valentina Nardi, Yvonne Li, Hersh Gupta, Andrew Cherniack, Mathew L. Meyerson, James M. Cleary, Jonathan A. Nowak, Brian M. Wolpin, and Andrew A. Aguirre
- Subjects
Cancer Research ,Oncology - Abstract
Approximately 8-10% of pancreatic cancers do not harbor mutations in KRAS. Understanding the unique molecular and clinical features of this subset of pancreatic cancer (PC) is important to guide patient stratification for clinical trials of molecularly targeted agents. To this end we investigated a cohort of 795 PC patients from Dana-Farber Cancer Institute who had undergone somatic genomic characterization with OncoPanel, a targeted next-generation sequencing panel with coverage of more than 400 cancer-associated genes. A total of 9.2% (73/795) of cases in our cohort were KRAS WT. The KRAS WT cohort was statistically enriched for MSI-H PC and acinar cell carcinomas (p = 0.0035, p < 0.0001 respectively). Actionable alterations in alternative MAPK drivers were identified in 44% (32/73) of KRAS WT cases. BRAF alterations accounted for 56% (18/32) of detected alternative MAPK drivers, the majority of which (72%) were Class II which exhibit dimer-dependent constitutive activity. Receptor Tyrosine Kinase (RTK) fusion events in BRAF, NTRK1, NRG1, NTRK3, ROS1, and FGFR2 accounted for 25% (8/32) of detected alternative MAPK drivers in KRAS WT tumors. BRAF in-frame deletions showed increased sensitivity to dual pan-RAF and MEK inhibition in organoid models and one patient with a ROS1 fusion received prolonged clinical benefit from targeted therapy. In addition to alternative MAPK drivers, mutations in GNAS (p = 0.0014) and ARID2 (p = 0.045) were significantly enriched in KRAS WT PC, whereas TP53 mutations were significantly less frequent in KRAS WT cases. Interestingly, although not statistically significant, rates of mutation in the other canonical tumor suppressor genes (CDKN2A, SMAD4) were also lower in KRAS WT PC. Clinically, KRAS Mutant (MUT) PC were associated with a decreased overall survival (OS) compared to the KRAS WT cohort [median OS 17.5 vs 24.0 months, HR 1.38, p = 0.036], however this relationship was no longer significant after accounting for other clinical factors. For patients with KRAS WT PC, those with SMAD4 alterations had a significantly decreased OS (HR 6.24, p < 0.001), whereas presence of TP53 or CDKN2A mutations had no significant impact. Lastly, we found that KRAS WT PC was associated with a younger age of onset. Interestingly, we noted that KRAS WT PC patients with a younger age of onset had tumors with few oncogenic alterations whereas no such association was seen in KRAS MUT patients. Validation of this finding in a separate dataset is required and is currently ongoing. In summary, our clinical and genomic characterization of KRAS WT PC identifies a high prevalence of alternative MAPK drivers that are amenable to targeted therapies. Our cohort also recapitulates the previously reported clinical characteristics of KRAS WT PC and identifies the presence of SMAD4 alterations as significantly associated with decreased overall survival in KRAS WT PC. Additional analysis from multiple sources will be critical to risk stratify these patients further and to validate age-related findings. Citation Format: Harshabad Singh, Rachel B. Keller, Kevin S. Kapner, Julien Dilly, Srivatsan Raghavan, Chen Yuan, Eizabeth Cohen, Michael Tolstorukov, Emma Hill, Elizabeth Andrews, Lauren K. Brais, Annacarolina Da Silva, Kimberly Perez, Douglas A. Rubinson, Benjamin L. Schlechter, Michael H. Rosenthal, Jason L. Hornick, Valentina Nardi, Yvonne Li, Hersh Gupta, Andrew Cherniack, Mathew L. Meyerson, James M. Cleary, Jonathan A. Nowak, Brian M. Wolpin, Andrew A. Aguirre. Clinical-genomic analysis of KRAS wild-type pancreatic cancer confirms alternative targetable drivers and provides insight for age and risk related clinical stratification [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer; 2022 Sep 13-16; Boston, MA. Philadelphia (PA): AACR; Cancer Res 2022;82(22 Suppl):Abstract nr A001.
- Published
- 2022
35. RNF43 G659fs is an oncogenic colorectal cancer mutation and sensitizes tumor cells to PI3K/mTOR inhibition
- Author
-
Lishan Fang, Dane Ford-Roshon, Max Russo, Casey O’Brien, Xiaozhe Xiong, Carino Gurjao, Maximilien Grandclaudon, Srivatsan Raghavan, Steven M. Corsello, Steven A. Carr, Namrata D. Udeshi, James Berstler, Ewa Sicinska, Kimmie Ng, and Marios Giannakis
- Subjects
Phosphatidylinositol 3-Kinases ,Multidisciplinary ,Cell Line, Tumor ,TOR Serine-Threonine Kinases ,Ubiquitin-Protein Ligases ,Mutation ,General Physics and Astronomy ,Humans ,General Chemistry ,Colorectal Neoplasms ,General Biochemistry, Genetics and Molecular Biology - Abstract
The RNF43_p.G659fs mutation occurs frequently in colorectal cancer, but its function remains poorly understood and there are no specific therapies directed against this alteration. In this study, we find that RNF43_p.G659fs promotes cell growth independent of Wnt signaling. We perform a drug repurposing library screen and discover that cells with RNF43_p.G659 mutations are selectively killed by inhibition of PI3K signaling. PI3K/mTOR inhibitors yield promising antitumor activity in RNF43659mut isogenic cell lines and xenograft models, as well as in patient-derived organoids harboring RNF43_p.G659fs mutations. We find that RNF43659mut binds p85 leading to increased PI3K signaling through p85 ubiquitination and degradation. Additionally, RNA-sequencing of RNF43659mut isogenic cells reveals decreased interferon response gene expression, that is reversed by PI3K/mTOR inhibition, suggesting that RNF43659mut may alter tumor immunity. Our findings suggest a therapeutic application for PI3K/mTOR inhibitors in treating RNF43_p.G659fs mutant cancers.
- Published
- 2021
36. Synthetic Lethal Interaction of SHOC2 Depletion with MEK Inhibition in RAS-Driven Cancers
- Author
-
Rosy Liao, Joshua Pan, Raymond W.S. Ng, Federica Piccioni, Jonathan P. Rennhack, Ari J. Firestone, William C. Hahn, Amy Goodale, Diego Almanza, Andrew J. Aguirre, John A. Bachman, Emma Pailler, David E. Root, Srivatsan Raghavan, Tina L. Yuan, Mukta Bagul, Nathanael S. Gray, Alfredo Gonzalez, Yenarae Lee, Jason J. Kwon, Katherine H. Walsh, Nancy Dumont, Timothy L. Bosse, Behnam Nabet, Francisca Vazquez, Jeff Settleman, Aviad Tsherniak, Joshua M. Dempster, Adrija J. Navarro, Maneesha Thaker, and Rita Sulahian
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Combination therapy ,MAP Kinase Signaling System ,Regulator ,Mice, SCID ,General Biochemistry, Genetics and Molecular Biology ,Receptor tyrosine kinase ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,Neoplasms ,Animals ,Humans ,Protein kinase A ,Protein Kinase Inhibitors ,lcsh:QH301-705.5 ,Cell Proliferation ,Mice, Hairless ,biology ,Effector ,Chemistry ,Intracellular Signaling Peptides and Proteins ,HCT116 Cells ,030104 developmental biology ,lcsh:Biology (General) ,Cell culture ,A549 Cells ,Cancer cell ,Cancer research ,biology.protein ,ras Proteins ,Mitogen-Activated Protein Kinases ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Summary: The mitogen-activated protein kinase (MAPK) pathway is a critical effector of oncogenic RAS signaling, and MAPK pathway inhibition may be an effective combination treatment strategy. We performed genome-scale loss-of-function CRISPR-Cas9 screens in the presence of a MEK1/2 inhibitor (MEKi) in KRAS-mutant pancreatic and lung cancer cell lines and identified genes that cooperate with MEK inhibition. While we observed heterogeneity in genetic modifiers of MEKi sensitivity across cell lines, several recurrent classes of synthetic lethal vulnerabilities emerged at the pathway level. Multiple members of receptor tyrosine kinase (RTK)-RAS-MAPK pathways scored as sensitizers to MEKi. In particular, we demonstrate that knockout, suppression, or degradation of SHOC2, a positive regulator of MAPK signaling, specifically cooperated with MEK inhibition to impair proliferation in RAS-driven cancer cells. The depletion of SHOC2 disrupted survival pathways triggered by feedback RTK signaling in response to MEK inhibition. Thus, these findings nominate SHOC2 as a potential target for combination therapy. : Sulahian, Kwon, and Walsh et al. performed several loss-of-function CRISPR-Cas9 screens in KRAS-mutant cancer cells treated with a MEK inhibitor and define the landscape of modifiers of MEK inhibitor sensitivity while highlighting that SHOC2 is a potent synthetic lethal target that serves as a critical signaling node to mediate MAP kinase pathway reactivation upon MEK inhibition. Keywords: Ras, KRAS, MEK inhibitor, synthetic lethal, SHOC2, CRISPR-Cas9 screen
- Published
- 2019
37. WRN helicase is a synthetic lethal target in microsatellite unstable cancers
- Author
-
Gad Getz, James M. McFarland, Aviad Tsherniak, Francisca Vazquez, Benjamin Gaeta, Yosef E. Maruvka, Marios Giannakis, Mirazul Islam, Monica Schenone, Edmond M. Chan, Annie Apffel, Sandy Chang, Paula Keskula, Alfredo Gonzalez, Jesse S. Boehm, Maria Alimova, Justine S. McPartlan, Tsukasa Shibue, Srivatsan Raghavan, Jean-Bernard Lazaro, Peili Gu, Yang Liu, Yanxi Zhang, Elizaveta Reznichenko, Lisa Leung, Jatin Roper, Tianxia Li, David E. Root, Raymond W.S. Ng, Emma A. Roberts, Alan D. D'Andrea, Jie Bin Liu, Syed O. Ali, Todd R. Golub, Zachary D. Nagel, Mahmoud Ghandi, Cortt G. Piett, Adam J. Bass, Nancy Dumont, Yuen-Yi Tseng, and Rebecca Deasy
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Werner Syndrome Helicase ,DNA repair ,Apoptosis ,Article ,03 medical and health sciences ,0302 clinical medicine ,RNA interference ,Cell Line, Tumor ,Neoplasms ,medicine ,Humans ,Gene silencing ,DNA Breaks, Double-Stranded ,Polymerase ,Multidisciplinary ,Models, Genetic ,biology ,nutritional and metabolic diseases ,Helicase ,Microsatellite instability ,Cell Cycle Checkpoints ,medicine.disease ,3. Good health ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Microsatellite Instability ,RNA Interference ,DNA mismatch repair ,CRISPR-Cas Systems ,Tumor Suppressor Protein p53 ,Synthetic Lethal Mutations ,Homologous recombination ,Microsatellite Repeats - Abstract
Synthetic lethality-an interaction between two genetic events through which the co-occurrence of these two genetic events leads to cell death, but each event alone does not-can be exploited for cancer therapeutics1. DNA repair processes represent attractive synthetic lethal targets, because many cancers exhibit an impairment of a DNA repair pathway, which can lead to dependence on specific repair proteins2. The success of poly(ADP-ribose) polymerase 1 (PARP-1) inhibitors in cancers with deficiencies in homologous recombination highlights the potential of this approach3. Hypothesizing that other DNA repair defects would give rise to synthetic lethal relationships, we queried dependencies in cancers with microsatellite instability (MSI), which results from deficient DNA mismatch repair. Here we analysed data from large-scale silencing screens using CRISPR-Cas9-mediated knockout and RNA interference, and found that the RecQ DNA helicase WRN was selectively essential in MSI models in vitro and in vivo, yet dispensable in models of cancers that are microsatellite stable. Depletion of WRN induced double-stranded DNA breaks and promoted apoptosis and cell cycle arrest selectively in MSI models. MSI cancer models required the helicase activity of WRN, but not its exonuclease activity. These findings show that WRN is a synthetic lethal vulnerability and promising drug target for MSI cancers.
- Published
- 2019
38. Sustainable dimethyl ether recycle system for regenerating super absorbent polymers
- Author
-
Srivatsan Raghavan
- Published
- 2020
39. Real-time Genomic Characterization of Advanced Pancreatic Cancer to Enable Precision Medicine
- Author
-
Emily E. Van Seventer, Ana Babic, Jeffrey W. Miller, Deborah Knoerzer, Marvin Ryou, Heather A. Shahzade, Nadine Jackson McCleary, Jaegil Kim, Douglas A. Rubinson, Mehlika Hazar-Rethinam, Rebecca J. Nagy, Kristin Anderka, David A. Tuveson, Robert J. Mayer, Scott L. Carter, Leona A. Doyle, Nelly Oliver, Nicholas D. Camarda, Kimmie Ng, Matthew B. Yurgelun, Lauren K. Brais, Levi A. Garraway, Jeffrey A. Meyerhardt, Arezou A. Ghazani, Richard A. Moffitt, Stuart G. Silverman, Thomas E. Clancy, Srivatsan Raghavan, Annacarolina da Silva, Brandon Nadres, James M. Cleary, Andrew J. Aguirre, Kunal Jajoo, Kelly P. Burke, Michael H. Rosenthal, Emma Reilly, Kimberly Perez, Jonathan A. Nowak, Charles S. Fuchs, Dean Welsch, Jen Jen Yeh, Matthew H. Kulke, Marisa W. Welch, William C. Hahn, Anuj K. Patel, Ryan B. Corcoran, Karla Helvie, Paul B. Shyn, Gad Getz, Nikhil Wagle, Dorisanne Y. Ragon, Lori Marini, Geoffrey I. Shapiro, Janet E. Murphy, Brian M. Wolpin, Richard B. Lanman, Devin McCabe, Jason L. Hornick, Bruce E. Johnson, Ewa Sicinska, and Joseph P. St. Pierre
- Subjects
Adult ,Male ,0301 basic medicine ,DNA Repair ,MAP Kinase Signaling System ,DNA repair ,Germline ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,Pancreatic cancer ,Exome Sequencing ,Carcinoma ,medicine ,Humans ,Gene Regulatory Networks ,Clinical significance ,Precision Medicine ,Homologous Recombination ,Germ-Line Mutation ,Exome sequencing ,Aged ,Aged, 80 and over ,Sequence Analysis, RNA ,business.industry ,Gene Expression Profiling ,Genetic Variation ,Genomics ,Middle Aged ,medicine.disease ,Precision medicine ,Gene Expression Regulation, Neoplastic ,Pancreatic Neoplasms ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Female ,business ,Carcinoma, Pancreatic Ductal - Abstract
Clinically relevant subtypes exist for pancreatic ductal adenocarcinoma (PDAC), but molecular characterization is not yet standard in clinical care. We implemented a biopsy protocol to perform time-sensitive whole-exome sequencing and RNA sequencing for patients with advanced PDAC. Therapeutically relevant genomic alterations were identified in 48% (34/71) and pathogenic/likely pathogenic germline alterations in 18% (13/71) of patients. Overall, 30% (21/71) of enrolled patients experienced a change in clinical management as a result of genomic data. Twenty-six patients had germline and/or somatic alterations in DNA-damage repair genes, and 5 additional patients had mutational signatures of homologous recombination deficiency but no identified causal genomic alteration. Two patients had oncogenic in-frame BRAF deletions, and we report the first clinical evidence that this alteration confers sensitivity to MAPK pathway inhibition. Moreover, we identified tumor/stroma gene expression signatures with clinical relevance. Collectively, these data demonstrate the feasibility and value of real-time genomic characterization of advanced PDAC. Significance: Molecular analyses of metastatic PDAC tumors are challenging due to the heterogeneous cellular composition of biopsy specimens and rapid progression of the disease. Using an integrated multidisciplinary biopsy program, we demonstrate that real-time genomic characterization of advanced PDAC can identify clinically relevant alterations that inform management of this difficult disease. Cancer Discov; 8(9); 1096–111. ©2018 AACR. See related commentary by Collisson, p. 1062. This article is highlighted in the In This Issue feature, p. 1047
- Published
- 2018
40. Microenvironment drives cell state, plasticity, and drug response in pancreatic cancer
- Author
-
Srivatsan Raghavan, Peter S. Winter, Andrew W. Navia, Hannah L. Williams, Alan DenAdel, Kristen E. Lowder, Jennyfer Galvez-Reyes, Radha L. Kalekar, Nolawit Mulugeta, Kevin S. Kapner, Manisha S. Raghavan, Ashir A. Borah, Nuo Liu, Sara A. Väyrynen, Andressa Dias Costa, Raymond W.S. Ng, Junning Wang, Emma K. Hill, Dorisanne Y. Ragon, Lauren K. Brais, Alex M. Jaeger, Liam F. Spurr, Yvonne Y. Li, Andrew D. Cherniack, Matthew A. Booker, Elizabeth F. Cohen, Michael Y. Tolstorukov, Isaac Wakiro, Asaf Rotem, Bruce E. Johnson, James M. McFarland, Ewa T. Sicinska, Tyler E. Jacks, Ryan J. Sullivan, Geoffrey I. Shapiro, Thomas E. Clancy, Kimberly Perez, Douglas A. Rubinson, Kimmie Ng, James M. Cleary, Lorin Crawford, Scott R. Manalis, Jonathan A. Nowak, Brian M. Wolpin, William C. Hahn, Andrew J. Aguirre, and Alex K. Shalek
- Subjects
Adult ,Male ,Middle Aged ,General Biochemistry, Genetics and Molecular Biology ,Gene Expression Regulation, Neoplastic ,Pancreatic Neoplasms ,Cell Line, Tumor ,Biomarkers, Tumor ,Tumor Microenvironment ,Humans ,Female ,Single-Cell Analysis ,Aged ,Carcinoma, Pancreatic Ductal - Abstract
Prognostically relevant RNA expression states exist in pancreatic ductal adenocarcinoma (PDAC), but our understanding of their drivers, stability, and relationship to therapeutic response is limited. To examine these attributes systematically, we profiled metastatic biopsies and matched organoid models at single-cell resolution. In vivo, we identify a new intermediate PDAC transcriptional cell state and uncover distinct site- and state-specific tumor microenvironments (TMEs). Benchmarking models against this reference map, we reveal strong culture-specific biases in cancer cell transcriptional state representation driven by altered TME signals. We restore expression state heterogeneity by adding back in vivo-relevant factors and show plasticity in culture models. Further, we prove that non-genetic modulation of cell state can strongly influence drug responses, uncovering state-specific vulnerabilities. This work provides a broadly applicable framework for aligning cell states across in vivo and ex vivo settings, identifying drivers of transcriptional plasticity and manipulating cell state to target associated vulnerabilities.
- Published
- 2021
41. Dewatering of super absorbent polymers: Alternatives to thermal desorption by liquid phase extraction using dimethyl ether
- Author
-
Srivatsan Raghavan, Jose F. Alfaro, Dimitris Ioannis Collias, Cheng Chi Wu, Alexandra Pine, and Brian J. Love
- Subjects
Desiccant ,Economics and Econometrics ,Aqueous solution ,genetic structures ,Extraction (chemistry) ,0211 other engineering and technologies ,Aqueous two-phase system ,02 engineering and technology ,010501 environmental sciences ,medicine.disease ,01 natural sciences ,Dewatering ,eye diseases ,chemistry.chemical_compound ,Superabsorbent polymer ,Chemical engineering ,chemistry ,medicine ,Dimethyl ether ,021108 energy ,Dehydration ,Waste Management and Disposal ,0105 earth and related environmental sciences - Abstract
Abstract The high saturation potential of highly hygroscopic desiccants makes their drying potential reuse an energetically unfavorable challenge. Perhaps there are other ways beyond dehydration the extract saturated water from these structures. That was the premise of the proposed experiments in which an existing method was redesigned in which condensed Dimethyl Ether (DME) was used aqueous fluids from commercial grade superabsorbent polymers (SAPs) similar to those found in commercial diapers. It was hypothesized that liquid extraction could be energetically less intensive than thermally dehydrating the SAP. By toggling between pressures above and below 0.6 MPa at room temperature, using gravity assistance, and nitrogen gas as driving forces, the DME condenses and interacts with the saturated SAP. The system successfully dewatered both DI water and saline saturated SAP. Roughly 90% of deionized water and ~80% of artificial urine are extractable from SAP when the chamber was pressurized 3 times. The salts in the artificial urine also conveyed in the aqueous extract and not concentrated in the dehydrated SAP. Energy assessments linked with each DME loading consumed 12.6 kJ or ~5 kJ/g of water extracted, more than required for thermal evaporation. The dehydrated superabsorbent polymer structure appears morphologically different by microscopy, but it can re-swell up to 35 times its dry mass in water and 15 times its mass in artificial urine. Overall, aqueous phase extraction using condensed DME dewaters saturated superabsorbent polymers allowing their reuse as other forms of desiccants but it is less efficient than by dehydration.
- Published
- 2021
42. Abstract 960: RNF43 G659fs is an oncogenic mutation in colorectal cancer and sensitizes tumor cells to PI3K/mTOR inhibition
- Author
-
Max Russo, Namrata D. Udeshi, Kimmie Ng, Casey O’Brien, Marios Giannakis, Steven M. Corsello, Maximilien Grandclaudon, Carino Gurjao, Srivatsan Raghavan, Dane Ford-Roshon, Lishan Fang, Ewa Sicinska, and James Berstler
- Subjects
Cancer Research ,Mutation ,Cell growth ,Wnt signaling pathway ,Cancer ,Biology ,medicine.disease_cause ,medicine.disease ,Frameshift mutation ,Oncology ,Cancer research ,medicine ,Carcinogenesis ,Protein kinase B ,PI3K/AKT/mTOR pathway - Abstract
Introduction: RNF43 is a transmembrane E3 ubiquitin ligase and WNT signaling suppressor that is commonly mutated in colorectal cancer (CRC). A C-terminal RNF43 hotspot mutation, RNF43_G659fs, occurs in approximately 36% (55/151) of microsatellite-instability (MSI)-high CRCs, but its underlying mechanism and function remain poorly understood. This study investigated the functional role of RNF43_G659fs in order to evaluate potential novel therapeutic approaches for tumors harboring this mutation. Methods: Isogenic RNF43117mut and RNF43659mut cell line models were generated using the CRISPR/Cas9 system to evaluate CRC tumorigenesis and WNT dependency. RNF43659mut was screened with a novel high-throughput drug repurposing library that employed a set of 5363 small molecules to identify compounds capable of selectively inhibiting RNF43659mut cell growth. Small molecules that selectively killed the RNF43659mut cells were validated in organoid models. Proteomic analysis, RNA-Seq and gene set enrichment analysis (GSEA) were performed to characterize mechanistic interactions and related signaling pathways of RNF43659mut in CRC. Results: Unlike N-terminal RNF43 frameshift mutations, we observed that RNF43659mut conferred a growth advantage over RNF43WT cells independent of WNT signaling. Furthermore, RNF43659mut and RNF43WT exhibited differential drug responses in the high-throughput drug repurposing screen which revealed that RNF43659mut cells were vulnerable to PI3K/AKT/mTOR inhibitors, including BYL-719 (Alpelisib). Enhanced AKT and mTOR activation was observed in RNF43659mut cell and attenuated by BYL-719 treatment in a dose-dependent manner. These results were subsequently validated in patient-derived organoid models. Furthermore, immunoprecipitation and proteomic analysis revealed interactions between RNF43_G659fs and p85, a negative regulator of PI3K. We also demonstrated that the RNF43_G659fs mutant activated PI3K/AKT/mTOR signaling through binding and degradation of p85. Consistent with the role of PI3K in immunomodulation, our RNA-Seq results showed that the RNF43_G659fs mutation was positively related to NF-kB activation (Normalized Enrichment Score=1.842, p Conclusion: This study confirms that RNF43659mut is an essential driver mutation in CRC and provides evidence that patients harboring RNF43_G659fs-mutant tumors may respond favorably to PI3K inhibition. Citation Format: Lishan Fang, Dane Ford-Roshon, Max Russo, Casey O'Brien, Carino Gurjao, Maximilien Grandclaudon, Steven M. Corsello, Srivatsan Raghavan, Namrata Udeshi, James Berstler, Ewa Sicinska, Kimmie Ng, Marios Giannakis. RNF43 G659fs is an oncogenic mutation in colorectal cancer and sensitizes tumor cells to PI3K/mTOR inhibition [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 960.
- Published
- 2021
43. Engineered Microenvironments to Investigate Cellular Behavior.
- Author
-
Srivatsan Raghavan, Celeste M. Nelson, Nathan J. Sniadecki, Emerson A. Lim, and Christopher S. Chen
- Published
- 2006
- Full Text
- View/download PDF
44. Non-viral delivery of CRISPR/Cas9 complex using CRISPR-GPS nanocomplexes
- Author
-
Sangeeta N. Bhatia, Srivatsan Raghavan, Piyush K. Jain, Santosh R. Rananaware, Heather E. Fleming, Michelle Tai, Justin H. Lo, Apekshya Panda, and Marco Downing
- Subjects
Gene Editing ,Cas9 ,Computer science ,Gene Transfer Techniques ,Translation (biology) ,02 engineering and technology ,Computational biology ,010402 general chemistry ,021001 nanoscience & nanotechnology ,01 natural sciences ,Article ,0104 chemical sciences ,Genome editing ,CRISPR ,Humans ,General Materials Science ,Guide RNA ,CRISPR-Cas Systems ,0210 nano-technology ,Ribonucleoprotein ,Subgenomic mRNA ,HeLa Cells - Abstract
There is a critical need for the development of safe and efficient delivery technologies for CRISPR/Cas9 to advance translation of genome editing to the clinic. Non-viral methods that are simple, efficient, and completely based on biologically-derived materials could offer such potential. Here we report a simple and modular tandem peptide-based nanocomplex system with cell-targeting capacity that efficiently combines guide RNA (sgRNA) with Cas9 protein, and facilitates internalization of sgRNA/Cas9 ribonucleoprotein complexes to yield robust genome editing across multiple cell lines.
- Published
- 2019
45. TAS-120 Overcomes Resistance to ATP-Competitive FGFR Inhibitors in Patients with FGFR2 Fusion-Positive Intrahepatic Cholangiocarcinoma
- Author
-
Islam Baiev, Raul N. Uppot, Leah Y. Liu, Giulia Siravegna, Isobel J Fetter, Stephanie Reyes, Dejan Juric, Cristina R. Ferrone, Emily E. Van Seventer, David T. Ting, Sachie Otsuki, Ipsita Dey-Guha, Jochen K. Lennerz, William C. Hahn, Lei Shi, Krushna C. Patra, Ignaty Leschiner, Gad Getz, Heather A. Shahzade, Cyril H. Benes, Phuong Vu, Robin Kate Kelley, Liudmila Elagina, Vikram Deshpande, Hiroshi Hirai, James J. Harding, Alberto Bardelli, Ferran Fece de la Cruz, Andrew X. Zhu, A. John Iafrate, Raymond W.S. Ng, Nabeel Bardeesy, Takeshi Sagara, Kenneth K. Tanabe, Rona Yaeger, Supriya K. Saha, Srivatsan Raghavan, Brandon Nadres, Janet E. Murphy, Ryan B. Corcoran, Ronald S. Arellano, and Lipika Goyal
- Subjects
0301 basic medicine ,Male ,Fibroblast Growth Factor ,Oncogene Proteins, Fusion ,Cell ,Drug Resistance ,Drug resistance ,medicine.disease_cause ,Circulating Tumor DNA ,Cholangiocarcinoma ,0302 clinical medicine ,Adenosine Triphosphate ,Erdafitinib ,Medicine ,Tomography ,Cancer ,Oncogene Proteins ,Mutation ,Tumor ,Kinase ,Middle Aged ,X-Ray Computed ,medicine.anatomical_structure ,Oncology ,Fibroblast growth factor receptor ,030220 oncology & carcinogenesis ,embryonic structures ,Female ,Type 2 ,Receptor ,Signal Transduction ,musculoskeletal diseases ,Adult ,animal structures ,FGFR Inhibition ,Oncology and Carcinogenesis ,Article ,Cell Line ,03 medical and health sciences ,Structure-Activity Relationship ,Cell Line, Tumor ,Genetics ,Humans ,Receptor, Fibroblast Growth Factor, Type 2 ,Fusion ,Protein Kinase Inhibitors ,Aged ,business.industry ,Phenylurea Compounds ,030104 developmental biology ,Pyrimidines ,Tumor progression ,Drug Resistance, Neoplasm ,Cancer research ,Neoplasm ,business ,Tomography, X-Ray Computed - Abstract
ATP-competitive fibroblast growth factor receptor (FGFR) kinase inhibitors, including BGJ398 and Debio 1347, show antitumor activity in patients with intrahepatic cholangiocarcinoma (ICC) harboring activating FGFR2 gene fusions. Unfortunately, acquired resistance develops and is often associated with the emergence of secondary FGFR2 kinase domain mutations. Here, we report that the irreversible pan-FGFR inhibitor TAS-120 demonstrated efficacy in 4 patients with FGFR2 fusion–positive ICC who developed resistance to BGJ398 or Debio 1347. Examination of serial biopsies, circulating tumor DNA (ctDNA), and patient-derived ICC cells revealed that TAS-120 was active against multiple FGFR2 mutations conferring resistance to BGJ398 or Debio 1347. Functional assessment and modeling the clonal outgrowth of individual resistance mutations from polyclonal cell pools mirrored the resistance profiles observed clinically for each inhibitor. Our findings suggest that strategic sequencing of FGFR inhibitors, guided by serial biopsy and ctDNA analysis, may prolong the duration of benefit from FGFR inhibition in patients with FGFR2 fusion–positive ICC.Significance:ATP-competitive FGFR inhibitors (BGJ398, Debio 1347) show efficacy in FGFR2-altered ICC; however, acquired FGFR2 kinase domain mutations cause drug resistance and tumor progression. We demonstrate that the irreversible FGFR inhibitor TAS-120 provides clinical benefit in patients with resistance to BGJ398 or Debio 1347 and overcomes several FGFR2 mutations in ICC models.This article is highlighted in the In This Issue feature, p. 983
- Published
- 2019
46. Abstract PR-006: Spatially resolved, single cell assessment of pancreatic ductal adenocarcinoma expression subtypes reveals mixed and hybrid basal-classical marker expression with prognostic significance and discrete spatial localization
- Author
-
Albert C. Koong, Andressa Dias Costa, Sara A. Väyrynen, Lauren K. Brais, Jonathan A. Nowak, Hannah Williams, Emma Reilly, Brian M. Wolpin, Alek K. Shalek, Srivatsan Raghavan, Jinming Zhang, Andrew J. Aguirre, David C. Linehan, Chen Yuan, Peter S. Winter, Margaret M. Kozak, Richard F. Dunne, Douglas A. Rubinson, Daniel T. Chang, Mai Chan Lau, Vicente Morales-Oyarvide, William C. Hahn, Aram F. Hezel, and Kevin S. Kapner
- Subjects
Cancer Research ,medicine.diagnostic_test ,Cell ,Cancer ,Biology ,medicine.disease ,Immunofluorescence ,Subtyping ,Basal (phylogenetics) ,medicine.anatomical_structure ,Oncology ,Pancreatic cancer ,medicine ,Cancer research ,Adenocarcinoma ,Progressive disease - Abstract
Background: Pancreatic adenocarcinoma (PDAC) is a rapidly progressive disease, with few molecular markers that stratify patients for survival time or treatment response. Bulk transcriptional analyses of PDAC have identified two major transcriptional subtypes, classical and basal, the latter of which is associated with worse overall survival and limited sensitivity to chemotherapy. However, spatial localization and classical-basal heterogeneity are incompletely understood at the individual cell level within a bulk tumor. Design: We built and validated a multiplex immunofluorescence assay that employs three classical and two basal subtype markers to measure subtype composition at spatially resolved, single cell resolution in a multi-institutional cohort of formalin-fixed paraffin-embedded primary pancreatic cancer resection specimens. Using digital image analysis, supervised machine learning, and Cox proportional hazards regression, we analyzed protein-level classical-basal subtype landscape, cell localization, and outcome associations. Results: Subtyping data were successfully generated for 1.2 million tumor cells across 290 resected tumor specimens (median 3579 cells/tumor). Tumor level analysis of cellular composition revealed that 88% of tumors are “mixed”, harboring any number of both classical and basal cells, whereas tumors composed only of classical or basal cells represented 11% and 1.4% of cases, respectively. In addition to pure classical and basal cells, we identified “hybrid cells” co-expressing basal and classical markers in 79% of cases, ranging in abundance from Citation Format: Hannah L. Williams, Jinming Zhang, Srivatsan Raghavan, Peter S. Winter, Kevin Kapner, Sara Vayrynen, Andressa Dias Costa, Chen Yuan, Mai Chan Lau, Vicente Morales-Oyarvide, Douglas Rubinson, Lauren Brais, Emma Reilly, Margaret Kozak, David Linehan, Richard Dunne, Daniel Chang, Albert Koong, Aram Hezel, William C. Hahn, Alek K. Shalek, Andrew J. Aguirre, Jonathan A. Nowak, Brian M Wolpin. Spatially resolved, single cell assessment of pancreatic ductal adenocarcinoma expression subtypes reveals mixed and hybrid basal-classical marker expression with prognostic significance and discrete spatial localization [abstract]. In: Proceedings of the AACR Virtual Special Conference on Pancreatic Cancer; 2020 Sep 29-30. Philadelphia (PA): AACR; Cancer Res 2020;80(22 Suppl):Abstract nr PR-006.
- Published
- 2020
47. Abstract PO-058: Transcriptional subtype-specific microenvironmental crosstalk and tumor cell plasticity in metastatic pancreatic cancer
- Author
-
Nolawit Mulugeta, Lauren K. Brais, James M. Cleary, Kristen E. Lowder, Brian M. Wolpin, Jonathan A. Nowak, Radha L. Kalekar, Emma Reilly, Andrew J. Aguirre, Alex K. Shalek, Junning Wang, William C. Hahn, Alan DenAdel, Srivatsan Raghavan, Manisha S. Raghavan, Jennyfer Galvez-Reyes, Dorisanne Y. Ragon, Alex M. Jaeger, Peter S. Winter, Lorin Crawford, Hannah Williams, Sara A. Väyrynen, Ashir A. Borah, Andressa Dias Costa, and Andrew W. Navia
- Subjects
Cancer Research ,education.field_of_study ,Cell ,Mesenchymal stem cell ,Population ,Biology ,medicine.disease ,Malignancy ,Phenotype ,medicine.anatomical_structure ,Oncology ,Pancreatic cancer ,Organoid ,medicine ,Cancer research ,education ,Progenitor - Abstract
Metastatic pancreatic ductal adenocarcinoma (PDAC) is an aggressive and lethal malignancy with few therapeutic options. Tumor transcriptional state is a strong predictor of clinical outcome in PDAC, with two primary cell states, basal-like and classical, identified by bulk transcriptional profiling. Basal-like tumors carry a worse prognosis, but the mechanisms underlying this survival difference, the degree of cellular heterogeneity within a given tumor, and the subtype-specific contributions from the local immune microenvironment are not well understood. In addition, there are ongoing efforts to use patient-derived organoid models as functional surrogates for an individual patient’s disease, but the degree to which patient transcriptional phenotypes are preserved in their matched organoid models remains unclear. Here, we describe a pipeline that enables both direct characterization of the liver metastatic niche via single-cell RNA-sequencing and functional assessment of PDAC tumor biology in patient-matched organoid models. Starting from core needle biopsies of metastatic PDAC lesions, we applied this approach to profile 22 patient samples and their matched organoid models using single-cell RNA-sequencing with Seq-Well. We demonstrate significant heterogeneity at the single-cell level across the basal-like to classical transcriptional spectrum. Basal-like cells expressed more mesenchymal and stem-like features, while classical cells expressed features of epithelial and pancreatic progenitor transcriptional programs. A population of “hybrid” malignant cells co-expressed markers of both basal-like and classical states, suggesting that these phenotypes lie on a continuum rather than as discrete entities. Microenvironmental composition also differed by subtype across T/NK and macrophage populations. Specifically, basal-like tumors exhibited tumor cell crosstalk with specific macrophage subsets, while classical tumors harbored greater immune infiltration and a relatively pro-angiogenic microenvironment, raising important considerations for subtype-specific microenvironmental directed therapy. Finally, we found that matched organoids exhibited transcriptional drift along the basal-like to classical axis relative to their parent tumors, with evidence for selection against basal-like phenotypes in vitro. However, tumor cells in organoid culture exhibited remarkable plasticity and could recover in vivo basal-like phenotypes in response to changes in their growth conditions. Taken together, our work provides a framework for the analysis of human cancers and their matched models using single-cell methods to dissect tumor-intrinsic and extrinsic contributions, and reveals novel insights into the transcriptional heterogeneity and plasticity of PDAC. Citation Format: Srivatsan Raghavan, Peter S. Winter, Andrew W. Navia, Hannah L. Williams, Alan DenAdel, Radha L. Kalekar, Jennyfer Galvez-Reyes, Kristen E. Lowder, Nolawit Mulugeta, Manisha S. Raghavan, Ashir A. Borah, Sara A. Vayrynen, Andressa Dias Costa, Junning Wang, Emma Reilly, Dorisanne Y. Ragon, Lauren K. Brais, Alex M. Jaeger, James M. Cleary, Lorin Crawford, Jonathan A. Nowak, Brian M. Wolpin, William C. Hahn, Andrew J. Aguirre, Alex K. Shalek. Transcriptional subtype-specific microenvironmental crosstalk and tumor cell plasticity in metastatic pancreatic cancer [abstract]. In: Proceedings of the AACR Virtual Special Conference on Pancreatic Cancer; 2020 Sep 29-30. Philadelphia (PA): AACR; Cancer Res 2020;80(22 Suppl):Abstract nr PO-058.
- Published
- 2020
48. Altered RNA Splicing by Mutant p53 Activates Oncogenic RAS Signaling in Pancreatic Cancer
- Author
-
Luisa F. Escobar-Hoyos, Peter Bailey, Karen Bai, Andrew V. Biankin, Srivatsan Raghavan, Daniel Dominguez, Daniel Kümmel, Andrew J. Aguirre, Grant A. Goda, Lisa H. Apken, Robert K. Bradley, Fong Cheng Pan, Simon J. Hogg, Cristian D. Cruz, Nicolas Lecomte, Barry S. Taylor, Joseph Saglimbeni, Paul Ogrodowski, Sruthi Babu, Kenneth R. Shroyer, Chun-Hao Pan, Hana Cho, Alex Penson, Brian M. Wolpin, Olivera Grbovic-Huezo, Scott W. Lowe, Renhe Luo, Yu-Jui Ho, Alessandro Pastore, John P. Morris, Andrea Oeckinghaus, David K. Chang, Andrea Ventura, Channing J. Der, Jerry P. Melchor, Sharon A. Lawrence, Steven D. Leach, Gokce Askan, Ram Kannan, Rohit Singh, Omar Abdel-Wahab, Direna Alonso-Curbelo, G. Aaron Hobbs, and Jonathan Bermeo
- Subjects
0301 basic medicine ,Cancer Research ,Spliceosome ,endocrine system diseases ,RNA Splicing ,Mutant ,Regulator ,Kaplan-Meier Estimate ,Biology ,medicine.disease_cause ,Article ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Cell Line, Tumor ,Pancreatic cancer ,medicine ,Animals ,Humans ,neoplasms ,Cells, Cultured ,Mice, Knockout ,Cancer ,Cell Biology ,medicine.disease ,Xenograft Model Antitumor Assays ,digestive system diseases ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,Pancreatic Neoplasms ,RNAi Therapeutics ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,RNA splicing ,Cancer research ,Female ,KRAS ,Tumor Suppressor Protein p53 ,Carcinoma, Pancreatic Ductal ,Signal Transduction - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is driven by co-existing mutations in KRAS and TP53. However, how these mutations collaborate to promote this cancer is unknown. Here, we uncover sequence-specific changes in RNA splicing enforced by mutant p53 which enhance KRAS activity. Mutant p53 increases expression of splicing regulator hnRNPK to promote inclusion of cytosine-rich exons within GTPase-activating proteins (GAPs), negative regulators of RAS family members. Mutant p53-enforced GAP isoforms lose cell membrane association, leading to heightened KRAS activity. Preventing cytosine-rich exon inclusion in mutant KRAS/p53 PDACs decreases tumor growth. Moreover, mutant p53 PDACs are sensitized to inhibition of splicing via spliceosome inhibitors. These data provide insight into co-enrichment of KRAS and p53 mutations and therapeutics targeting this mechanism in PDAC.
- Published
- 2020
49. Abstract PR02: Matched metastatic pancreatic ductal adenocarcinoma biopsies and organoid models reveal tumor cell transcriptional plasticity and subtype-specific microenvironmental crosstalk
- Author
-
Alex K. Shalek, Junning Wang, Emma Reilly, Lorin Crawford, Radha L. Kalekar, Brian M. Wolpin, Andrew W. Navia, William C. Hahn, Nolawit Mulugeta, Lauren K. Brais, James M. McFarland, Hannah Williams, Jonathan A. Nowak, Ashir A. Borah, Jennyfer Galvez-Reyes, Srivatsan Raghavan, Alan DenAdel, Peter S. Winter, James M. Cleary, Kristen E. Lowder, Andrew J. Aguirre, and Manisha S. Raghavan
- Subjects
Cancer Research ,Tumor microenvironment ,Cell ,Disease ,Biology ,Phenotype ,Pathogenesis ,Crosstalk (biology) ,Immune system ,medicine.anatomical_structure ,Oncology ,Organoid ,Cancer research ,medicine - Abstract
The majority of patients with pancreatic ductal adenocarcinoma (PDAC) present with metastatic disease at diagnosis and have median survival times of less than 12 months. Recent studies have demonstrated that PDAC tumors with distinct transcriptional signatures are associated with different clinical outcomes, and that the tumor microenvironment may contribute to PDAC pathogenesis. In parallel, there are ongoing efforts to understand if patient-derived organoid models can be used as functional surrogates for an individual patient’s disease. However, it remains unclear if patient transcriptional phenotypes are preserved in their matched organoid models. Here, we describe a pipeline that permits both direct characterization of the PDAC liver metastatic niche via single-cell RNA-sequencing and functional assessment of PDAC tumor biology in patient-matched organoid models. Starting from core needle biopsies of metastatic PDAC lesions containing 50-100k viable cells, we simultaneously perform (1) low-input single-cell RNA-sequencing using Seq-Well and (2) three-dimensional organoid culture generation. We have applied this approach to profile 21 patients and their matched early passage organoid models. Our pipeline yields high-quality single-cell measurements across diverse cell types—both tumor and nontumor stromal—enabling a principled dissection of tumor intrinsic and extrinsic factors. Evaluation of clinically relevant transcriptional signatures (e.g., basal-like vs. classical) revealed extensive heterogeneity at the single-cell level and identified new, hybrid expression states. We also observed evidence of significant subtype-specific crosstalk between immune populations and tumor cells—specifically between T cells and tumor cells originating from basal-like tumors. Serial sampling at different stages of treatment revealed transcriptional shifts in tumor cells suggestive of significant plasticity. We similarly found that organoids derived from basal-like tumors exhibited considerable plasticity in vitro and had decreased fitness in standard organoid culture conditions, suggesting the need for distinct environments to support specific transcriptional subtypes. Overall, our approach provides actionable insights into the heterogeneity and plasticity of human PDAC, as well as a pipeline and framework for the analysis of PDAC and other cancers. This abstract is also being presented as Poster A50. Citation Format: Peter S. Winter, Srivatsan Raghavan, Andrew W. Navia, Hannah Williams, Jennyfer Galvez-Reyes, Radha Kalekar, Ashir Borah, Alan DenAdel, Manisha Raghavan, Kristen Lowder, Nolawit Mulugeta, Junning Wang, Emma Reilly, Lauren Brais, Lorin Crawford, James McFarland, James M. Cleary, Jonathan Nowak, Brian M. Wolpin, Andrew J. Aguirre, William C. Hahn, Alex K. Shalek. Matched metastatic pancreatic ductal adenocarcinoma biopsies and organoid models reveal tumor cell transcriptional plasticity and subtype-specific microenvironmental crosstalk [abstract]. In: Proceedings of the AACR Special Conference on the Evolving Landscape of Cancer Modeling; 2020 Mar 2-5; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2020;80(11 Suppl):Abstract nr PR02.
- Published
- 2020
50. Phase Ib study of gemcitabine, nab-paclitaxel, and ficlatuzumab in patients with advanced pancreatic cancer
- Author
-
Brian M. Wolpin, Basil Bakir, Lawrence S. Blaszkowsky, Colin D. Weekes, Jeffrey A. Meyerhardt, James M. Cleary, Andrew J. Aguirre, Matthew B. Yurgelun, Marios Giannakis, Lipika Goyal, Koushik K. Das, Kimberly Perez, Thomas A. Abrams, Jason R. Pitarresi, Srivatsan Raghavan, Anil K. Rustgi, Douglas A. Rubinson, Michael N. Needle, Danielle Stonely, and Peter C. Enzinger
- Subjects
0301 basic medicine ,Gene isoform ,Cancer Research ,business.industry ,medicine.disease ,medicine.disease_cause ,Gemcitabine ,03 medical and health sciences ,030104 developmental biology ,Oncology ,Ficlatuzumab ,Pancreatic cancer ,medicine ,Cancer research ,Pancreatitis ,Hepatocyte growth factor ,Carcinogenesis ,business ,Transcription factor ,medicine.drug - Abstract
693 Background: Paired-related homeodomain transcription factor 1 (Prrx1) isoforms are involved in pancreatic development, pancreatitis, and carcinogenesis. Hepatocyte growth factor (HGF) is a transcriptional target of Prrx1b. Ficlatuzumab is a recombinant humanized HGF antibody, that neutralizes HGF/c-Met binding and HGF-induced c-Met phosphorylation. In preclinical pancreatic ductal adenocarcinoma (PDAC) models, inhibition of Prrx1b-HGF signaling using ficlatuzumab and gemcitabine reduced primary tumor volume and eliminated metastatic disease. Methods: Patients (pts) with previously-untreated metastatic PDAC enrolled in a phase Ib dose escalation study with 3+3 design and two dose cohorts of ficlatuzumab (10mg/kg and 20mg/kg) administered intravenously every other week with gemcitabine (G; 1000mg/m2) and nab-paclitaxel (A; 125mg/m2) given 3 weeks on and 1 week off. This was followed by an expansion phase at the maximally tolerated dose (MTD) of the combination. Results: 24 pts (sex, 12M:12F; median age, 69 years [range, 51-82 years]) were enrolled. No dose-limiting toxicities were identified in the phase 1b (N = 6 pts) and ficlatuzumab at 20mg/kg with GA was advanced to the expansion phase (N = 18 pts). By RECISTv1.1 in the full study population, 7 (29%) pts had partial response, 15 (63%) had stable disease, and 2 (8%) could not be evaluated. Median progression-free survival was 8 months (range, 3-16 months), 4 pts are still on study treatment. The primary toxicities attributed to ficlatuzumab included hypoalbuminemia (grade 3, 21%; any grade, 91%) and edema (grade 3, 8%; any grade, 91%). Nine (38%) of the 24 pts discontinued study treatment due to these toxicities prior to disease progression. Conclusions: The combination of ficlatuzumab with gemcitabine and nab-paclitaxel is associated with durable treatment responses but also significant hypoalbuminemia and edema that may impair treatment tolerability. Serial blood samples were collected for circulating HGF measurements, and mandatory pretreatment biopsies were collected for tumor c-MET pathway activation and 3D organoid culture drug sensitivity testing. Clinical trial information: NCT03316599.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.