23 results on '"Emily Beirne"'
Search Results
2. Supplemental Table 1 from Multiplexed Evaluation of Microdosed Antineoplastic Agents In Situ in the Tumor Microenvironment of Patients with Soft Tissue Sarcoma
- Author
-
Robert G. Maki, Richard A. Klinghoffer, Micah Ellison, Joyoti Dey, Jason Frazier, Emily Beirne, Kimberly H.W. Sottero, Marc O. Grenley, Jessica A. Bertout, William Kerwin, Daniel C. Ramirez, Mee-Young Lee, Jessica L. Davis, Matthew J. Thompson, Seth M. Pollack, Howard J. Goodman, Gary B. Deutsch, and Kenneth R. Gundle
- Abstract
Table of microinjected drugs
- Published
- 2023
- Full Text
- View/download PDF
3. Supplemental Figure 5 from Multiplexed Evaluation of Microdosed Antineoplastic Agents In Situ in the Tumor Microenvironment of Patients with Soft Tissue Sarcoma
- Author
-
Robert G. Maki, Richard A. Klinghoffer, Micah Ellison, Joyoti Dey, Jason Frazier, Emily Beirne, Kimberly H.W. Sottero, Marc O. Grenley, Jessica A. Bertout, William Kerwin, Daniel C. Ramirez, Mee-Young Lee, Jessica L. Davis, Matthew J. Thompson, Seth M. Pollack, Howard J. Goodman, Gary B. Deutsch, and Kenneth R. Gundle
- Abstract
Microinjection of olaratumab does not inhibit PDGFRα, ERK, or S6 phosphorylation.
- Published
- 2023
- Full Text
- View/download PDF
4. Supplemental Table 2 from Multiplexed Evaluation of Microdosed Antineoplastic Agents In Situ in the Tumor Microenvironment of Patients with Soft Tissue Sarcoma
- Author
-
Robert G. Maki, Richard A. Klinghoffer, Micah Ellison, Joyoti Dey, Jason Frazier, Emily Beirne, Kimberly H.W. Sottero, Marc O. Grenley, Jessica A. Bertout, William Kerwin, Daniel C. Ramirez, Mee-Young Lee, Jessica L. Davis, Matthew J. Thompson, Seth M. Pollack, Howard J. Goodman, Gary B. Deutsch, and Kenneth R. Gundle
- Abstract
All adverse events reported on the study
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Legend from Multiplexed Evaluation of Microdosed Antineoplastic Agents In Situ in the Tumor Microenvironment of Patients with Soft Tissue Sarcoma
- Author
-
Robert G. Maki, Richard A. Klinghoffer, Micah Ellison, Joyoti Dey, Jason Frazier, Emily Beirne, Kimberly H.W. Sottero, Marc O. Grenley, Jessica A. Bertout, William Kerwin, Daniel C. Ramirez, Mee-Young Lee, Jessica L. Davis, Matthew J. Thompson, Seth M. Pollack, Howard J. Goodman, Gary B. Deutsch, and Kenneth R. Gundle
- Abstract
Supplementary Legend
- Published
- 2023
- Full Text
- View/download PDF
6. Supplemental Figure 3 from Multiplexed Evaluation of Microdosed Antineoplastic Agents In Situ in the Tumor Microenvironment of Patients with Soft Tissue Sarcoma
- Author
-
Robert G. Maki, Richard A. Klinghoffer, Micah Ellison, Joyoti Dey, Jason Frazier, Emily Beirne, Kimberly H.W. Sottero, Marc O. Grenley, Jessica A. Bertout, William Kerwin, Daniel C. Ramirez, Mee-Young Lee, Jessica L. Davis, Matthew J. Thompson, Seth M. Pollack, Howard J. Goodman, Gary B. Deutsch, and Kenneth R. Gundle
- Abstract
Distinct drug induced phenotypes and apoptotic responses specific to drug mechanism of action are seen at sites of CIVO microinjection.
- Published
- 2023
- Full Text
- View/download PDF
7. Data from Multiplexed Evaluation of Microdosed Antineoplastic Agents In Situ in the Tumor Microenvironment of Patients with Soft Tissue Sarcoma
- Author
-
Robert G. Maki, Richard A. Klinghoffer, Micah Ellison, Joyoti Dey, Jason Frazier, Emily Beirne, Kimberly H.W. Sottero, Marc O. Grenley, Jessica A. Bertout, William Kerwin, Daniel C. Ramirez, Mee-Young Lee, Jessica L. Davis, Matthew J. Thompson, Seth M. Pollack, Howard J. Goodman, Gary B. Deutsch, and Kenneth R. Gundle
- Abstract
Purpose:A persistent issue in cancer drug development is the discordance between robust antitumor drug activity observed in laboratory models and the limited benefit frequently observed when patients are treated with the same agents in clinical trials. Difficulties in accurately modeling the complexities of human tumors may underlie this problem. To address this issue, we developed Comparative In Vivo Oncology (CIVO), which enables in situ investigation of multiple microdosed drugs simultaneously in a patient's tumor. This study was designed to test CIVO's safety and feasibility in patients with soft tissue sarcoma (STS).Patients and Methods:We conducted a single arm, prospective, 13-patient pilot study. Patients scheduled for incisional biopsy or tumor resection were CIVO-injected 1 to 3 days prior to surgery. Saline or microdoses of anticancer agents were percutaneously injected into the tumor in a columnar fashion through each of eight needles. Following excision, drug responses were evaluated in the injected tissue.Results:The primary objective was met, establishing CIVO's feasibility and safety. Device-related adverse events were limited to transient grade 1 nonserious events. In addition, biomarker evaluation of localized tumor response to CIVO microinjected drugs by IHC or with NanoString GeoMx Digital Spatial Profiler demonstrated consistency with known mechanisms of action of each drug, impact on the tumor microenvironment, and historic clinical activity.Conclusions:These results are an advance toward use of CIVO as a translational research tool for early evaluation of investigational agents and drug combinations in a novel approach to phase 0 trials.See related commentary by Sleijfer and Lolkema, p. 3897
- Published
- 2023
- Full Text
- View/download PDF
8. Supplemental Figure 2 from Multiplexed Evaluation of Microdosed Antineoplastic Agents In Situ in the Tumor Microenvironment of Patients with Soft Tissue Sarcoma
- Author
-
Robert G. Maki, Richard A. Klinghoffer, Micah Ellison, Joyoti Dey, Jason Frazier, Emily Beirne, Kimberly H.W. Sottero, Marc O. Grenley, Jessica A. Bertout, William Kerwin, Daniel C. Ramirez, Mee-Young Lee, Jessica L. Davis, Matthew J. Thompson, Seth M. Pollack, Howard J. Goodman, Gary B. Deutsch, and Kenneth R. Gundle
- Abstract
Representative images of tumors from patients who received radiation treatment.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplemental Table 3 from Multiplexed Evaluation of Microdosed Antineoplastic Agents In Situ in the Tumor Microenvironment of Patients with Soft Tissue Sarcoma
- Author
-
Robert G. Maki, Richard A. Klinghoffer, Micah Ellison, Joyoti Dey, Jason Frazier, Emily Beirne, Kimberly H.W. Sottero, Marc O. Grenley, Jessica A. Bertout, William Kerwin, Daniel C. Ramirez, Mee-Young Lee, Jessica L. Davis, Matthew J. Thompson, Seth M. Pollack, Howard J. Goodman, Gary B. Deutsch, and Kenneth R. Gundle
- Abstract
Device performance
- Published
- 2023
- Full Text
- View/download PDF
10. Supplemental Figure 1 from Multiplexed Evaluation of Microdosed Antineoplastic Agents In Situ in the Tumor Microenvironment of Patients with Soft Tissue Sarcoma
- Author
-
Robert G. Maki, Richard A. Klinghoffer, Micah Ellison, Joyoti Dey, Jason Frazier, Emily Beirne, Kimberly H.W. Sottero, Marc O. Grenley, Jessica A. Bertout, William Kerwin, Daniel C. Ramirez, Mee-Young Lee, Jessica L. Davis, Matthew J. Thompson, Seth M. Pollack, Howard J. Goodman, Gary B. Deutsch, and Kenneth R. Gundle
- Abstract
Clinical Trial Design
- Published
- 2023
- Full Text
- View/download PDF
11. Supplemental Figure 4 from Multiplexed Evaluation of Microdosed Antineoplastic Agents In Situ in the Tumor Microenvironment of Patients with Soft Tissue Sarcoma
- Author
-
Robert G. Maki, Richard A. Klinghoffer, Micah Ellison, Joyoti Dey, Jason Frazier, Emily Beirne, Kimberly H.W. Sottero, Marc O. Grenley, Jessica A. Bertout, William Kerwin, Daniel C. Ramirez, Mee-Young Lee, Jessica L. Davis, Matthew J. Thompson, Seth M. Pollack, Howard J. Goodman, Gary B. Deutsch, and Kenneth R. Gundle
- Abstract
Increased phosphorylation of PDGFR� and downstream effectors in an undifferentiated pleomorphic sarcoma displaying a lack of apoptotic response to microdosed doxorubicin.
- Published
- 2023
- Full Text
- View/download PDF
12. 569 A CIVO intratumor microdose Phase 0 Trial of subasumstat (TAK-981) in combination with cetuximab or avelumab reveals Type 1 Interferon induction and immune activation in head and neck cancer patients
- Author
-
Jeffrey Houlton, Daniel Clayburgh, Ryan Li, Nathan Schauer, Marc Grenley, Connor Burns, Angela Merrell, Jason Frazier, Jonathan Derry, Emily Beirne, Wendy Jenkins, Allison Berger, and Richard Klinghoffer
- Published
- 2022
- Full Text
- View/download PDF
13. Multiplexed Evaluation of Microdosed Antineoplastic Agents In Situ in the Tumor Microenvironment of Patients with Soft Tissue Sarcoma
- Author
-
Jessica A. Bertout, Kimberly H.W. Sottero, Richard A. Klinghoffer, Jason Frazier, Howard J. Goodman, Matthew J. Thompson, Gary B. Deutsch, Daniel C. Ramirez, Joyoti Dey, Marc Grenley, Mee Young Lee, Kenneth R. Gundle, William S. Kerwin, Micah Ellison, Jessica L. Davis, Robert G. Maki, Seth M. Pollack, and Emily Beirne
- Subjects
0301 basic medicine ,Oncology ,Drug ,Cancer Research ,Tumor microenvironment ,medicine.medical_specialty ,business.industry ,media_common.quotation_subject ,Soft tissue sarcoma ,Translational research ,medicine.disease ,Clinical trial ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,In vivo ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Biomarker (medicine) ,Adverse effect ,business ,media_common - Abstract
Purpose: A persistent issue in cancer drug development is the discordance between robust antitumor drug activity observed in laboratory models and the limited benefit frequently observed when patients are treated with the same agents in clinical trials. Difficulties in accurately modeling the complexities of human tumors may underlie this problem. To address this issue, we developed Comparative In Vivo Oncology (CIVO), which enables in situ investigation of multiple microdosed drugs simultaneously in a patient's tumor. This study was designed to test CIVO's safety and feasibility in patients with soft tissue sarcoma (STS). Patients and Methods: We conducted a single arm, prospective, 13-patient pilot study. Patients scheduled for incisional biopsy or tumor resection were CIVO-injected 1 to 3 days prior to surgery. Saline or microdoses of anticancer agents were percutaneously injected into the tumor in a columnar fashion through each of eight needles. Following excision, drug responses were evaluated in the injected tissue. Results: The primary objective was met, establishing CIVO's feasibility and safety. Device-related adverse events were limited to transient grade 1 nonserious events. In addition, biomarker evaluation of localized tumor response to CIVO microinjected drugs by IHC or with NanoString GeoMx Digital Spatial Profiler demonstrated consistency with known mechanisms of action of each drug, impact on the tumor microenvironment, and historic clinical activity. Conclusions: These results are an advance toward use of CIVO as a translational research tool for early evaluation of investigational agents and drug combinations in a novel approach to phase 0 trials. See related commentary by Sleijfer and Lolkema, p. 3897
- Published
- 2020
- Full Text
- View/download PDF
14. Abstract CT139: Intratumoral (IT) microdosing of the investigational SUMOylation Inhibitor TAK-981 in a phase 0 CIVO trial demonstrates the reactivation of type I Interferon (IFN1) signaling in head and neck squamous cell carcinoma (HNSCC)
- Author
-
Jeffrey Houlton, Harrison Cash, Haodong Xu, Paul L. Swiecicki, Keith Casper, Steven B. Chinn, Daniel R. Clayburgh, Ryan J. Li, Robert J. Christian, Aaron Halfpenny, Annemieke van Zante, Beryl A. Hatton, Kimberly Sottero, Marc O. Grenley, Connor Burns, Jason Frazier, Jonathan Derry, Gloria Kung, Emily Beirne, Nathan J. Schauer, Atticus Turner, Wendy Jenkins, Kirsten Anderson, Richard A. Klinghoffer, Dennis Huszar, Allison Berger, and Karuppiah Kannan
- Subjects
Cancer Research ,Oncology - Abstract
Purpose/Objectives: The human tumor microenvironment (TME) has a dramatic impact on cancer prognosis and therapeutic response, but accurate models of the native TME do not exist. The Comparative In Vivo Oncology (CIVO) platform was developed as a means to assess the effect of investigational agents on the native TME in a Phase 0 microdosing study. CIVO was clinically validated using approved agents and is used for the first time here to assess the impact of an investigational agent - the SUMOylation inhibitor TAK-981 - on the native human TME in HNSCC. Materials/Methods: Eligible subjects have a confirmed HNSCC diagnosis, ECOG 0-2, and planned surgical resection. Injectable tumors were at the primary site or within cervical lymph nodes but had to be surface-accessible and ≥ 2cm. TAK-981 or control microdoses were simultaneously administered via a CIVO device and co-injected with a fluorescent tracking marker for injection site identification and visualization. Tumors were resected 24 or 72 hours after injection, processed, and then analyzed at a central site. Multiplexed biomarker staining and molecular profiling via GeoMx Digital Spatial Profiling were performed to capture pharmacodynamic responses in the native TME. Results: As of January 2022, 8 subjects provided informed consent and were enrolled, and no adverse events associated with the injection procedure or microdoses have been reported. Biomarker analysis demonstrated TAK-981 distribution around the injection site accompanied by reduction of SUMOylation. Dose-dependent elevation of IFN1 signaling was also observed in TAK-981-exposed areas within the TME. Elevated IFN1 signaling was accompanied by TME reconfiguration, with increased macrophage M1 polarization and activation of dendritic cells, NK cells, and CD8+ T cells. TAK-981 exposure was also associated with upregulation of CXCL10, PD-L1, and an IFNγ gene expression signature predictive of response to immune checkpoint blockade. Conclusion: IT microdosing with CIVO provided early insights into complex functional responses induced by the investigational agent TAK-981 that can only be accurately evaluated in the intact, native TME of a patient’s tumor. SUMO pathway inhibition in HNSCC tumors following TAK-981 exposure led to functional activation of multiple immune cell types, effectively shifting the local TME toward an inflamed “hot” state, highlighting TAK-981’s potential as an immune stimulating agent for treating patients with solid tumors. These data were generated while TAK-981 was still in Phase I dose escalation trials (via IV administration), highlighting CIVO’s ability to safely study investigational agents. Further evaluation of TAK-981 alone and in combination with other agents is ongoing in this Phase 0 CIVO microdosing trial. Citation Format: Jeffrey Houlton, Harrison Cash, Haodong Xu, Paul L. Swiecicki, Keith Casper, Steven B. Chinn, Daniel R. Clayburgh, Ryan J. Li, Robert J. Christian, Aaron Halfpenny, Annemieke van Zante, Beryl A. Hatton, Kimberly Sottero, Marc O. Grenley, Connor Burns, Jason Frazier, Jonathan Derry, Gloria Kung, Emily Beirne, Nathan J. Schauer, Atticus Turner, Wendy Jenkins, Kirsten Anderson, Richard A. Klinghoffer, Dennis Huszar, Allison Berger, Karuppiah Kannan. Intratumoral (IT) microdosing of the investigational SUMOylation Inhibitor TAK-981 in a phase 0 CIVO trial demonstrates the reactivation of type I Interferon (IFN1) signaling in head and neck squamous cell carcinoma (HNSCC) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr CT139.
- Published
- 2022
- Full Text
- View/download PDF
15. Multiplexed Evaluation of Microdosed Antineoplastic Agents
- Author
-
Kenneth R, Gundle, Gary B, Deutsch, Howard J, Goodman, Seth M, Pollack, Matthew J, Thompson, Jessica L, Davis, Mee-Young, Lee, Daniel C, Ramirez, William, Kerwin, Jessica A, Bertout, Marc O, Grenley, Kimberly H W, Sottero, Emily, Beirne, Jason, Frazier, Joyoti, Dey, Micah, Ellison, Richard A, Klinghoffer, and Robert G, Maki
- Subjects
Tumor Microenvironment ,Humans ,Antineoplastic Agents ,Pilot Projects ,Sarcoma ,Prospective Studies - Abstract
A persistent issue in cancer drug development is the discordance between robust antitumor drug activity observed in laboratory models and the limited benefit frequently observed when patients are treated with the same agents in clinical trials. Difficulties in accurately modeling the complexities of human tumors may underlie this problem. To address this issue, we developed ComparativeWe conducted a single arm, prospective, 13-patient pilot study. Patients scheduled for incisional biopsy or tumor resection were CIVO-injected 1 to 3 days prior to surgery. Saline or microdoses of anticancer agents were percutaneously injected into the tumor in a columnar fashion through each of eight needles. Following excision, drug responses were evaluated in the injected tissue.The primary objective was met, establishing CIVO's feasibility and safety. Device-related adverse events were limited to transient grade 1 nonserious events. In addition, biomarker evaluation of localized tumor response to CIVO microinjected drugs by IHC or with NanoString GeoMx Digital Spatial Profiler demonstrated consistency with known mechanisms of action of each drug, impact on the tumor microenvironment, and historic clinical activity.These results are an advance toward use of CIVO as a translational research tool for early evaluation of investigational agents and drug combinations in a novel approach to phase 0 trials.
- Published
- 2020
16. Voruciclib, a clinical stage oral CDK9 inhibitor, represses MCL-1 and sensitizes high-risk Diffuse Large B-cell Lymphoma to BCL2 inhibition
- Author
-
Kate C. Gillespie, Thomas L. Deckwerth, Richard A. Klinghoffer, Joseph Casalini, Joyoti Dey, Edward F. Kleinman, Sally Ditzler, Marc Grenley, Emily Beirne, Chantel P. Dixon, Angela Merrell, Connor Burns, and William S. Kerwin
- Subjects
0301 basic medicine ,Combination therapy ,medicine.medical_treatment ,lcsh:Medicine ,Antineoplastic Agents ,Apoptosis ,Article ,Targeted therapy ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,immune system diseases ,Cell Line, Tumor ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Benzopyrans ,lcsh:Science ,Chemotherapy ,Sulfonamides ,Multidisciplinary ,business.industry ,Venetoclax ,lcsh:R ,Germinal center ,Drug Synergism ,medicine.disease ,Bridged Bicyclo Compounds, Heterocyclic ,Lymphoma ,030104 developmental biology ,chemistry ,Gene Expression Regulation ,Proto-Oncogene Proteins c-bcl-2 ,030220 oncology & carcinogenesis ,Imino Furanoses ,Cancer research ,Myeloid Cell Leukemia Sequence 1 Protein ,lcsh:Q ,Lymphoma, Large B-Cell, Diffuse ,business ,Diffuse large B-cell lymphoma - Abstract
Aberrant regulation of BCL-2 family members enables evasion of apoptosis and tumor resistance to chemotherapy. BCL-2 and functionally redundant counterpart, MCL-1, are frequently over-expressed in high-risk diffuse large B-cell lymphoma (DLBCL). While clinical inhibition of BCL-2 has been achieved with the BH3 mimetic venetoclax, anti-tumor efficacy is limited by compensatory induction of MCL-1. Voruciclib, an orally bioavailable clinical stage CDK-selective inhibitor, potently blocks CDK9, the transcriptional regulator of MCL-1. Here, we demonstrate that voruciclib represses MCL-1 protein expression in preclinical models of DLBCL. When combined with venetoclax in vivo, voruciclib leads to model-dependent tumor cell apoptosis and tumor growth inhibition. Strongest responses were observed in two models representing high-risk activated B-cell (ABC) DLBCL, while no response was observed in a third ABC model, and intermediate responses were observed in two models of germinal center B-cell like (GCB) DLBCL. Given the range of responses, we show that CIVO, a multiplexed tumor micro-dosing technology, represents a viable functional precision medicine approach for differentiating responders from non-responders to BCL-2/MCL-1 targeted therapy. These findings suggest that the combination of voruciclib and venetoclax holds promise as a novel, exclusively oral combination therapy for a subset of high-risk DLBCL patients.
- Published
- 2017
- Full Text
- View/download PDF
17. Establishment and characterization of a canine soft tissue sarcoma patient-derived xenograft model
- Author
-
Derek Thirstrup, Jason Frazier, Ilona Tretyak, Richard A. Klinghoffer, Sue E. Knoblaugh, Joseph Casalini, J. G. Ward, Emily Beirne, C. D. Tripp, and Sally Ditzler
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,General Veterinary ,business.industry ,Soft tissue sarcoma ,Canine Soft Tissue Sarcoma ,medicine.disease ,Gemcitabine ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Docetaxel ,030220 oncology & carcinogenesis ,Systemic administration ,Cancer research ,medicine ,Immunohistochemistry ,Doxorubicin ,Sarcoma ,business ,medicine.drug - Abstract
Spontaneously occurring soft tissue sarcoma (STS) is relatively common in canine cancer patients. Because of the similarities to human disease, canine STSs are a valuable and readily available resource for the study of new therapeutics. In this study, a canine patient-derived xenograft (PDX) model, CDX-STS2, was established. The CDX-STS2 model was engrafted and expanded for systemic administration studies with chemotherapeutic agents commonly used to treat STS, including doxorubicin, docetaxel and gemcitabine. Immunohistochemistry for drug-specific biomarkers and tumour growth measurement revealed tumour sensitivity to doxorubicin and docetaxel, whereas gemcitabine had no effect on tumour growth. Although many human PDX tumour models have been established, relatively few canine PDX models have been reported to date. CDX-STS2 represents a new STS PDX research model of canine origin that will be useful in bridging preclinical research with clinical studies of STS in pet dogs.
- Published
- 2016
- Full Text
- View/download PDF
18. Multidrug Analyses in Patients Distinguish Efficacious Cancer Agents Based on Both Tumor Cell Killing and Immunomodulation
- Author
-
Ilona Tretyak, Jessica A. Bertout, Andy Keener, Korashon L. Watts, Sarah Gillings, Karri A. Meleo, Megan N. Breit, Alicia Moreno-Gonzalez, Jason Frazier, Sally Ditzler, Kevin Choy, Chelsea D. Tripp, Derek Thirstrup, Joey R. Casalini, Ravi K. Amaravadi, James A. Perry, Richard A. Klinghoffer, Emily Beirne, Vanessa Rizzo, Marc Grenley, William S. Kerwin, Chamisa L. Herrera, and James M. Olson
- Subjects
0301 basic medicine ,Drug ,Cancer Research ,media_common.quotation_subject ,Antineoplastic Agents ,Drug resistance ,Article ,Immunomodulation ,03 medical and health sciences ,0302 clinical medicine ,Dogs ,In vivo ,Cell Line, Tumor ,Neoplasms ,Medicine ,Animals ,Humans ,Doxorubicin ,Precision Medicine ,media_common ,business.industry ,Cancer ,medicine.disease ,Drug Resistance, Multiple ,Pre-clinical development ,030104 developmental biology ,Oncology ,Drug development ,030220 oncology & carcinogenesis ,Cancer cell ,Immunology ,Cancer research ,business ,medicine.drug - Abstract
The vision of a precision medicine–guided approach to novel cancer drug development is challenged by high intratumor heterogeneity and interpatient diversity. This complexity is rarely modeled accurately during preclinical drug development, hampering predictions of clinical drug efficacy. To address this issue, we developed Comparative In Vivo Oncology (CIVO) arrayed microinjection technology to test tumor responsiveness to simultaneous microdoses of multiple drugs directly in a patient's tumor. Here, in a study of 18 canine patients with soft tissue sarcoma (STS), CIVO captured complex, patient-specific tumor responses encompassing both cancer cells and multiple immune infiltrates following localized exposure to different chemotherapy agents. CIVO also classified patient-specific tumor resistance to the most effective agent, doxorubicin, and further enabled assessment of a preclinical autophagy inhibitor, PS-1001, to reverse doxorubicin resistance. In a CIVO-identified subset of doxorubicin-resistant tumors, PS-1001 resulted in enhanced antitumor activity, increased infiltration of macrophages, and skewed this infiltrate toward M1 polarization. The ability to evaluate and cross-compare multiple drugs and drug combinations simultaneously in living tumors and across a diverse immunocompetent patient population may provide a foundation from which to make informed drug development decisions. This method also represents a viable functional approach to complement current precision oncology strategies. Cancer Res; 77(11); 2869–80. ©2017 AACR.
- Published
- 2017
19. Abstract 4136: Direct intratumoral microdosing via the CIVO® platform reveals anti-tumor immune responses induced by the SUMO inhibitor TAK-981
- Author
-
Joyoti Dey, Sally Ditzler, Connor Burns, Beryl A. Hatton, Vaishali Shinde, Dennis Huszar, Marc Grenley, James Garnsey, Richard A. Klinghoffer, Emily Beirne, and Angela Merrell
- Subjects
Cancer Research ,Tumor microenvironment ,biology ,business.industry ,Microdosing ,SUMO protein ,Acquired immune system ,Immune system ,Oncology ,Granzyme ,Interferon ,Cancer research ,biology.protein ,Medicine ,Cytotoxic T cell ,business ,medicine.drug - Abstract
The complex interplay between a drug, the tumor and the surrounding microenvironment is a critical determinant in how a patient will respond to their selected cancer therapy. Yet to date, this complex response has been difficult to evaluate prior to late stage clinical investigation. To enable a means for testing investigational agents earlier in the development process but still directly in human patients, in a way that limits the risk of adverse effects and provides an indication of efficacy, Presage Biosciences developed a technology called CIVO. This platform allows for simultaneous assessment of multiple drugs or drug combinations directly in a single solid tumor, in the context where the drugs are ultimately intended to be used, directly in the patient. Here we demonstrate the potential for using the CIVO platform in Phase 0 microdosing studies to detect complex responses to investigational agents. In this study, we used the CIVO platform to assess the impact of TAK-981 on the native tumor immune microenvironment of animal models. TAK-981 is a novel and selective small molecule inhibitor of the SUMOylation enzymatic cascade currently in Phase I clinical trials. SUMOylation is a reversible post-translational modification that regulates protein function by covalent attachment of a small ubiquitin-like modifier (SUMO) protein to protein substrates. TAK-981 was microinjected into tumors from a syngeneic mouse model of B cell lymphoma and responses assessed via immunohistochemistry and in situ hybridization following tumor resection. An early inflammatory response was evident by 24 hours, including the accumulation of neutrophils, inflammatory macrophages and a Type I interferon response. The chemokine IP10 was secreted around TAK-981 injection sites and was accompanied by the accumulation of cytotoxic T lymphocytes, likely recruited from the local tumor microenvironment. A localized cell death response was observed proximal to TAK-981 injection sites by 72 hours and was likely induced by the granzyme B-bearing cytotoxic T cells enriched at TAK-981 sites. Abscopal studies demonstrated that the local immune modulation induced by TAK-981 translated into an adaptive immune response. The results from this study were consistent with findings from systemically dosed in vivomouse efficacy studies carried out at Takeda, demonstrating that the local responses to agents microdosed intratumorally via CIVO are predictive of responses induced by systemic drug exposure. These studies highlight the unique capability of TAK-981 to promote antitumor immunity, which may be further evaluated using the CIVO platform in a Phase 0 trial in human solid tumor patients. Citation Format: Beryl A. Hatton, Marc Grenley, James Garnsey, Vaishali Shinde, Dennis Huszar, Connor Burns, Sally Ditzler, Angela Merrell, Joyoti Dey, Emily Beirne, Richard A. Klinghoffer. Direct intratumoral microdosing via the CIVO® platform reveals anti-tumor immune responses induced by the SUMO inhibitor TAK-981 [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 4136.
- Published
- 2019
- Full Text
- View/download PDF
20. Abstract 2155: High-plex spatial profiling analysis of multidrug CIVO microdose studies in cancer patients
- Author
-
Jason Frazier, Yan Liang, Gary B. Deutsch, Seth M. Pollack, Robert G. Maki, Joseph M. Beechem, Matthew Thompson, Kenneth R. Gundle, Jessica A. Bertout, Marc Grenley, Richard A. Klinghoffer, Emily Beirne, and Jingjing Gong
- Subjects
Cancer Research ,business.industry ,Microdosing ,RNA ,medicine.disease ,Oncology ,Drug development ,Aldesleukin ,Gene expression ,medicine ,Cancer research ,Doxorubicin ,Sarcoma ,Biomarker discovery ,business ,medicine.drug - Abstract
Background: CIVO microdosing studies performed in patient tumors in situ allow drug developers to assess localized tumor and microenvironment responses to multiple agents without having to expose patients to high systemic drug levels. By concentrating microdoses of multiple different drugs within a living tumor in situ, it is possible to compare tumor and immune responses in spatially resolved regions of the same tumor. This type of early phase (Phase 0) clinical study represents a new path for drug developers to gain insight into drug efficacy, tumor associated immune cell modulation, biomarker discovery and validation, and microenvironment interactions for new drugs earlier in the drug development process. Traditionally, analysis of FFPE samples from these microdosing studies involve routine immunohistochemistry, immunofluorescence, and in situ assays that reveal changes in protein and RNA expression. These assays offer tissue wide protein and gene expression information but have limited multiplexing capabilities and dynamic range, as well as rapidly consume precious trial samples. Novel technologies such as NanoString’s GeoMxTM Digital Spatial Profiler (DSP) enable high-plex spatially resolved analysis of proteins and RNA transcripts in single FFPE tissue sections. In this proof of principle study, we utilized GeoMxTM Digital Spatial Profiler for protein and RNA expression on single FFPE sections from patient sarcoma tumors that were microdosed with multiple FDA approved drugs. Methods: Single FFPE sections from microdosed patient tumor samples were IF stained for DNA and CD3 and whole slide imaged. 600-micron and 100-micron diameter regions of interests (ROIs) were selected for DSP analysis within drug and control microinjection sites. Imaging and barcode counts were performed using GeoMxTM DSP and nCounter systems. Results: DSP protein analysis highlighted phospho-S6 and phospho-ERK upregulation in response to doxorubicin compared to vehicle site. Additionally, ROIs sampled along the doxorubicin exposure gradient showed a dose-dependent reduction of phosphorylation of both S6 and ERK proteins. DSP RNA analysis revealed drug specific transcript regulation of multiple genes in microdosed tumors, including upregulation of chemokines CXCL9 and CXCL10 at sites of doxorubicin and aldesleukin injection but not to other chemotherapy agents. Conclusions: Early phase CIVO microdosing studies combined with high-plex DSP opens the door to generating multi-omics data for multiple microdosed drugs within small patient studies. Analysis of protein and RNA expression using DSP enabled collection of targeted region of interest high density data from single FFPE sections, conserving precious patient biopsy samples. Through continued expansion of the GeoMxTM DSP analyte panels, collecting an ever-increasing depth of protein and gene expression data is possible in Phase 0 CIVO microdosing studies. Citation Format: Gary B. Deutsch, Seth M. Pollack, Matthew J. Thompson, Kenneth R. Gundle, Jessica A. Bertout, Jason P. Frazier, Emily Beirne, Marc O. Grenley, JingJing Gong, Yan Liang, Joseph M. Beechem, Richard A. Klinghoffer, Robert G. Maki. High-plex spatial profiling analysis of multidrug CIVO microdose studies in cancer patients [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 2155.
- Published
- 2019
- Full Text
- View/download PDF
21. Abstract CT142: Multidrug analyses in cancer patients via intratumoral microdosing with CIVO: A functional approach to precision oncology
- Author
-
Marc Grenley, Matthew J. Thompson, Seth M. Pollack, Kimberly H.W. Sottero, Jessica A. Bertout, Richard A. Klinghoffer, Darin Davidson, and Emily Beirne
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Microdosing ,business.industry ,Precision oncology ,Internal medicine ,medicine ,Cancer ,business ,medicine.disease - Abstract
The vision of a precision medicine-guided approach to novel cancer drug development is challenged by high intra-tumor heterogeneity, interpatient diversity, and complex interactions between tumor cells and the surrounding microenvironment. Attempts to model this complexity through the use of ex vivo systems have failed to accurately represent tumor biology, and resulting predictions of clinical drug efficacy have met with limited success. The era of genomics, highlighted by remarkable advances in high-throughput next generation sequencing technology, has sparked hope that this issue can be solved through a precision medicine approach to cancer treatment: matching the right drugs to the right patients. However, despite a few noteworthy successes pairing targeted agents with oncogenic driver mutations, genome-based precision medicine has been limited by an incomplete understanding of the relationship between cancer mutations and drug responsiveness. To overcome these issues, we developed CIVO (Comparative In Vivo Oncology), an arrayed microinjection technology that evaluates tumor responsiveness to microdoses of multiple drugs simultaneously and directly in a patient's tumor. CIVO consists of a multidrug injection device, fluorescent tracking microspheres which accurately denote the position of each drug injection site, and automated image analysis tools capable of quantifying both tumor cell and microenvironmental biomarkers of response to local drug exposure. Here we present preliminary data on the first 4 patients from our clinical feasibility study in soft tissue sarcoma. No unexpected adverse events related to microdose injection were observed. Consistent with historical clinical data, front line agent doxorubicin induced localized increases in markers for DNA damage, tumor cell apoptosis, and macrophage infiltration, whereas gemcitabine did not induce any observable local responses. Doxorubicin also induced tumor cell responses that suggest potential mechanisms of resistance to single agent therapy, including upregulation of PDGFαR and ERK phosphorylation. Multidrug investigation via intratumoral microinjection with CIVO in patients with soft tissue sarcoma appears safe and feasible. The ability to evaluate and cross-compare multiple drugs and drug combinations simultaneously in living tumors across a diverse immune competent patient population provides an innovative and exciting functional approach to complement current precision oncology strategies and enables deep characterization of cancer biology in early drug development. Citation Format: Matthew J. Thompson, Darin J. Davidson, Jessica A. Bertout, Kimberly H. Sottero, Marc O. Grenley, Emily Beirne, Richard A. Klinghoffer, Seth M. Pollack. Multidrug analyses in cancer patients via intratumoral microdosing with CIVO: A functional approach to precision oncology [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr CT142.
- Published
- 2018
- Full Text
- View/download PDF
22. Multidrug analyses in cancer patients via intratumoral microdosing with CIVO: A microinjection technology for phase 0 drug investigation
- Author
-
Richard A. Klinghoffer, Emily Beirne, Jessica A. Bertout, Matthew J. Thompson, Darin Davidson, Kimberly H.W. Sottero, Marc Grenley, and Seth M. Pollack
- Subjects
Oncology ,Drug ,Cancer Research ,medicine.medical_specialty ,business.industry ,Microdosing ,media_common.quotation_subject ,Cancer drugs ,Cancer ,medicine.disease ,Internal medicine ,Medicine ,business ,human activities ,Microinjection ,media_common - Abstract
e23569Background: The vision of a precision medicine-guided approach to novel cancer drug development is challenged by high intra-tumor heterogeneity, interpatient diversity, and complex interactio...
- Published
- 2018
- Full Text
- View/download PDF
23. A technology platform to assess multiple cancer agents simultaneously within a patient's tumor
- Author
-
Richard A. Klinghoffer, Joseph Casalini, Emily J. Girard, Michael Carleton, Kyle Pedro, Thomas L. Deckwerth, Shelli M. Morris, Ilona Tretyak, Andrew D. Strand, Emily Beirne, James M. Olson, Sheng You, Derek Thirstrup, Oliver W. Press, Sally Ditzler, Korashon L. Watts, William S. Kerwin, Jason Frazier, Alicia Moreno-Gonzalez, Rajesh Chopra, Jessica A. Bertout, Joyoti Dey, Mandana Veiseh, Beryl A. Hatton, Ellen Filvaroff, Marc Grenley, Karri A. Meleo, and S. Bahram Bahrami
- Subjects
Drug ,Vincristine ,Lymphoma ,Prednisolone ,media_common.quotation_subject ,Mice, Nude ,Antineoplastic Agents ,Mice, SCID ,Drug resistance ,Pharmacology ,Article ,Mice ,chemistry.chemical_compound ,Dogs ,Drug Delivery Systems ,Mafosfamide ,In vivo ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Doxorubicin ,Cyclophosphamide ,media_common ,business.industry ,TOR Serine-Threonine Kinases ,Cancer ,General Medicine ,medicine.disease ,chemistry ,Drug Resistance, Neoplasm ,Cancer research ,Drug Monitoring ,Drug Screening Assays, Antitumor ,business ,Biomarkers ,Neoplasm Transplantation ,medicine.drug - Abstract
A fundamental problem in cancer drug development is that antitumor efficacy in preclinical cancer models does not translate faithfully to patient outcomes. Much of early cancer drug discovery is performed under in vitro conditions in cell-based models that poorly represent actual malignancies. To address this inconsistency, we have developed a technology platform called CIVO, which enables simultaneous assessment of up to eight drugs or drug combinations within a single solid tumor in vivo. The platform is currently designed for use in animal models of cancer and patients with superficial tumors but can be modified for investigation of deeper-seated malignancies. In xenograft lymphoma models, CIVO microinjection of well-characterized anticancer agents (vincristine, doxorubicin, mafosfamide, and prednisolone) induced spatially defined cellular changes around sites of drug exposure, specific to the known mechanisms of action of each drug. The observed localized responses predicted responses to systemically delivered drugs in animals. In pair-matched lymphoma models, CIVO correctly demonstrated tumor resistance to doxorubicin and vincristine and an unexpected enhanced sensitivity to mafosfamide in multidrug-resistant lymphomas compared with chemotherapy-naïve lymphomas. A CIVO-enabled in vivo screen of 97 approved oncology agents revealed a novel mTOR (mammalian target of rapamycin) pathway inhibitor that exhibits significantly increased tumor-killing activity in the drug-resistant setting compared with chemotherapy-naïve tumors. Finally, feasibility studies to assess the use of CIVO in human and canine patients demonstrated that microinjection of drugs is toxicity-sparing while inducing robust, easily tracked, drug-specific responses in autochthonous tumors, setting the stage for further application of this technology in clinical trials.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.