270 results on '"Gavin P Dunn"'
Search Results
52. Data from Circulating Immune Cell and Outcome Analysis from the Phase II Study of PD-L1 Blockade with Durvalumab for Newly Diagnosed and Recurrent Glioblastoma
- Author
-
David A. Reardon, Thomas Kaley, Raymond Y. Huang, Todd Creasy, Nicholas M. Durham, Melissa de los Reyes, Ralph Venhaus, Paul Schwarzenberger, Aileen Ryan, Toni Ricciardi, Andrew J. Park, Mary Macri, Patrick Y. Wen, Christina Holcroft, Julia Hayden, Sarah Gaffey, Elizabeth George, Elizabeth Flagg, Hui K. Gan, Timothy Cloughesy, Michael Lim, Gavin P. Dunn, Jennifer L. Clarke, Jorg Dietrich, Nathan Standifer, and Lakshmi Nayak
- Abstract
Purpose:PD-L1 is upregulated in glioblastoma and supports immunosuppression. We evaluated PD-L1 blockade with durvalumab among glioblastoma cohorts and investigated potential biomarkers.Patients and Methods:MGMT unmethylated newly diagnosed patients received radiotherapy plus durvalumab (cohort A; n = 40). Bevacizumab-naïve, recurrent patients received durvalumab alone (cohort B; n = 31) or in combination with standard bevacizumab (cohort B2; n = 33) or low-dose bevacizumab (cohort B3; n = 33). Bevacizumab-refractory patients received durvalumab plus bevacizumab (cohort C; n = 22). Primary endpoints were: OS-12 (A), PFS-6 (B, B2, B3), and OS-6 (C). Exploratory biomarkers included: a systematic, quantitative, and phenotypic evaluation of circulating immune cells; tumor mutational burden (TMB); and tumor immune activation signature (IAS).Results:No cohort achieved the primary efficacy endpoint. Outcome was comparable among recurrent, bevacizumab-naïve cohorts. No unexpected toxicities were observed. A widespread reduction of effector immune cell subsets was noted among recurrent patients compared with newly diagnosed patients that was partially due to dexamethasone use. A trend of increased CD8+Ki67+ T cells at day 15 was noted among patients who achieved the primary endpoint and were not on dexamethasone. Neither TMB nor IAS predicted outcome.Conclusions:Patients with recurrent glioblastoma have markedly lower baseline levels of multiple circulating immune cell subsets compared with newly diagnosed patients. An early increase in systemic Ki67+CD8+ cells may warrant further evaluation as a potential biomarker of therapeutic benefit among patients with glioblastoma undergoing checkpoint therapy. Dexamethasone decreased immune cell subsets. PD-L1 blockade and combination with standard or reduced dose bevacizumab was ineffective.
- Published
- 2023
53. Supplementary Figure 5 from Yap1 Mediates Trametinib Resistance in Head and Neck Squamous Cell Carcinomas
- Author
-
Ravindra Uppaluri, Obi L. Griffith, Ann Marie Egloff, Malachi Griffith, Douglas R. Adkins, Gavin P. Dunn, Tusar Giri, Ibrahim Ozgenc, Erica K. Barnell, Rachel Riley, Zachary L. Skidmore, Paul Zolkind, Jason Webb, Katie M. Campbell, and Tenny Mudianto
- Abstract
Supplementary Figure 5
- Published
- 2023
54. Supplementary Figure 7 from Neoadjuvant and Adjuvant Pembrolizumab in Resectable Locally Advanced, Human Papillomavirus–Unrelated Head and Neck Cancer: A Multicenter, Phase II Trial
- Author
-
Douglas R. Adkins, Obi L. Griffith, Malachi Griffith, Rebecca D. Chernock, Jay F. Piccirillo, Dorina Kallogjeri, Ian S. Hagemann, Scott J. Rodig, Mackenzie Daly, Hiram A. Gay, Wade Thorstad, Ana Lako, Evisa Gjini, Jonathan D. Schoenfeld, Robert Haddad, Jason Kass, Glenn J. Hanna, Matthew D. Stachler, Vickie Y. Jo, Liye Zhou, Rachel Riley, Tenny Mudianto, Trevor J. Pugh, Iulia Cirlan, Youstina Hanna, David T. Mulder, Tiantian Li, Tianxiang Lin, Nicholas C. Spies, Erica K. Barnell, Gavin P. Dunn, Peter Oppelt, Jessica Ley, Loren S. Michel, Patrik Pipkorn, Ryan Jackson, Jason T. Rich, Randal C. Paniello, Brian Nussenbaum, Zachary L. Skidmore, Paul Zolkind, Ann Marie Egloff, Katie M. Campbell, and Ravindra Uppaluri
- Abstract
Supplementary Figure
- Published
- 2023
55. Supplementary Data from Circulating Immune Cell and Outcome Analysis from the Phase II Study of PD-L1 Blockade with Durvalumab for Newly Diagnosed and Recurrent Glioblastoma
- Author
-
David A. Reardon, Thomas Kaley, Raymond Y. Huang, Todd Creasy, Nicholas M. Durham, Melissa de los Reyes, Ralph Venhaus, Paul Schwarzenberger, Aileen Ryan, Toni Ricciardi, Andrew J. Park, Mary Macri, Patrick Y. Wen, Christina Holcroft, Julia Hayden, Sarah Gaffey, Elizabeth George, Elizabeth Flagg, Hui K. Gan, Timothy Cloughesy, Michael Lim, Gavin P. Dunn, Jennifer L. Clarke, Jorg Dietrich, Nathan Standifer, and Lakshmi Nayak
- Abstract
Supplementary Data from Circulating Immune Cell and Outcome Analysis from the Phase II Study of PD-L1 Blockade with Durvalumab for Newly Diagnosed and Recurrent Glioblastoma
- Published
- 2023
56. Supplementary Appendix from Neoadjuvant and Adjuvant Pembrolizumab in Resectable Locally Advanced, Human Papillomavirus–Unrelated Head and Neck Cancer: A Multicenter, Phase II Trial
- Author
-
Douglas R. Adkins, Obi L. Griffith, Malachi Griffith, Rebecca D. Chernock, Jay F. Piccirillo, Dorina Kallogjeri, Ian S. Hagemann, Scott J. Rodig, Mackenzie Daly, Hiram A. Gay, Wade Thorstad, Ana Lako, Evisa Gjini, Jonathan D. Schoenfeld, Robert Haddad, Jason Kass, Glenn J. Hanna, Matthew D. Stachler, Vickie Y. Jo, Liye Zhou, Rachel Riley, Tenny Mudianto, Trevor J. Pugh, Iulia Cirlan, Youstina Hanna, David T. Mulder, Tiantian Li, Tianxiang Lin, Nicholas C. Spies, Erica K. Barnell, Gavin P. Dunn, Peter Oppelt, Jessica Ley, Loren S. Michel, Patrik Pipkorn, Ryan Jackson, Jason T. Rich, Randal C. Paniello, Brian Nussenbaum, Zachary L. Skidmore, Paul Zolkind, Ann Marie Egloff, Katie M. Campbell, and Ravindra Uppaluri
- Abstract
supplementary appendix
- Published
- 2023
57. Supplementary Figure 10 from Neoadjuvant and Adjuvant Pembrolizumab in Resectable Locally Advanced, Human Papillomavirus–Unrelated Head and Neck Cancer: A Multicenter, Phase II Trial
- Author
-
Douglas R. Adkins, Obi L. Griffith, Malachi Griffith, Rebecca D. Chernock, Jay F. Piccirillo, Dorina Kallogjeri, Ian S. Hagemann, Scott J. Rodig, Mackenzie Daly, Hiram A. Gay, Wade Thorstad, Ana Lako, Evisa Gjini, Jonathan D. Schoenfeld, Robert Haddad, Jason Kass, Glenn J. Hanna, Matthew D. Stachler, Vickie Y. Jo, Liye Zhou, Rachel Riley, Tenny Mudianto, Trevor J. Pugh, Iulia Cirlan, Youstina Hanna, David T. Mulder, Tiantian Li, Tianxiang Lin, Nicholas C. Spies, Erica K. Barnell, Gavin P. Dunn, Peter Oppelt, Jessica Ley, Loren S. Michel, Patrik Pipkorn, Ryan Jackson, Jason T. Rich, Randal C. Paniello, Brian Nussenbaum, Zachary L. Skidmore, Paul Zolkind, Ann Marie Egloff, Katie M. Campbell, and Ravindra Uppaluri
- Abstract
Supplementary Figure
- Published
- 2023
58. Supplementary tables from Yap1 Mediates Trametinib Resistance in Head and Neck Squamous Cell Carcinomas
- Author
-
Ravindra Uppaluri, Obi L. Griffith, Ann Marie Egloff, Malachi Griffith, Douglas R. Adkins, Gavin P. Dunn, Tusar Giri, Ibrahim Ozgenc, Erica K. Barnell, Rachel Riley, Zachary L. Skidmore, Paul Zolkind, Jason Webb, Katie M. Campbell, and Tenny Mudianto
- Abstract
Supplementary Tables
- Published
- 2023
59. Supplementary Figure 6 from Yap1 Mediates Trametinib Resistance in Head and Neck Squamous Cell Carcinomas
- Author
-
Ravindra Uppaluri, Obi L. Griffith, Ann Marie Egloff, Malachi Griffith, Douglas R. Adkins, Gavin P. Dunn, Tusar Giri, Ibrahim Ozgenc, Erica K. Barnell, Rachel Riley, Zachary L. Skidmore, Paul Zolkind, Jason Webb, Katie M. Campbell, and Tenny Mudianto
- Abstract
Supplementary Figure 6
- Published
- 2023
60. Supplementary Tables from Neoadjuvant and Adjuvant Pembrolizumab in Resectable Locally Advanced, Human Papillomavirus–Unrelated Head and Neck Cancer: A Multicenter, Phase II Trial
- Author
-
Douglas R. Adkins, Obi L. Griffith, Malachi Griffith, Rebecca D. Chernock, Jay F. Piccirillo, Dorina Kallogjeri, Ian S. Hagemann, Scott J. Rodig, Mackenzie Daly, Hiram A. Gay, Wade Thorstad, Ana Lako, Evisa Gjini, Jonathan D. Schoenfeld, Robert Haddad, Jason Kass, Glenn J. Hanna, Matthew D. Stachler, Vickie Y. Jo, Liye Zhou, Rachel Riley, Tenny Mudianto, Trevor J. Pugh, Iulia Cirlan, Youstina Hanna, David T. Mulder, Tiantian Li, Tianxiang Lin, Nicholas C. Spies, Erica K. Barnell, Gavin P. Dunn, Peter Oppelt, Jessica Ley, Loren S. Michel, Patrik Pipkorn, Ryan Jackson, Jason T. Rich, Randal C. Paniello, Brian Nussenbaum, Zachary L. Skidmore, Paul Zolkind, Ann Marie Egloff, Katie M. Campbell, and Ravindra Uppaluri
- Abstract
Supplementary tables
- Published
- 2023
61. Supplementary Data from Cancers from Novel Pole-Mutant Mouse Models Provide Insights into Polymerase-Mediated Hypermutagenesis and Immune Checkpoint Blockade
- Author
-
Uri Tabori, Zachary F. Pursell, Cynthia J. Guidos, Cynthia E. Hawkins, James G. Jackson, Zucai Suo, Eric Bouffet, Adam Shlien, Alberto Martin, Carol Durno, Oz Mordechai, Jonathan N. Byrd, Gavin P. Dunn, Tomasz Sarosiek, Andrea Seeley, Jagadeesh Ramdas, Xia Wang, Melyssa Aronson, Nuno M. Nunes, Victoria J. Forster, Jiil Chung, Dana Elshaer, Richard de Borja, Melissa Edwards, Robert Siddaway, Iram Siddiqui, Lazar Joksimovic, Ayse B. Ercan, Vivian S. Park, Nathan A. Ungerleider, Walter J. Zahurancik, Taylor Bridge, Sumedha Sudhaman, Miki S. Gams, Karl P. Hodel, and Melissa A. Galati
- Abstract
Supplemental Titles and Figure Legends
- Published
- 2023
62. Supplementary Figure 6 from Neoadjuvant and Adjuvant Pembrolizumab in Resectable Locally Advanced, Human Papillomavirus–Unrelated Head and Neck Cancer: A Multicenter, Phase II Trial
- Author
-
Douglas R. Adkins, Obi L. Griffith, Malachi Griffith, Rebecca D. Chernock, Jay F. Piccirillo, Dorina Kallogjeri, Ian S. Hagemann, Scott J. Rodig, Mackenzie Daly, Hiram A. Gay, Wade Thorstad, Ana Lako, Evisa Gjini, Jonathan D. Schoenfeld, Robert Haddad, Jason Kass, Glenn J. Hanna, Matthew D. Stachler, Vickie Y. Jo, Liye Zhou, Rachel Riley, Tenny Mudianto, Trevor J. Pugh, Iulia Cirlan, Youstina Hanna, David T. Mulder, Tiantian Li, Tianxiang Lin, Nicholas C. Spies, Erica K. Barnell, Gavin P. Dunn, Peter Oppelt, Jessica Ley, Loren S. Michel, Patrik Pipkorn, Ryan Jackson, Jason T. Rich, Randal C. Paniello, Brian Nussenbaum, Zachary L. Skidmore, Paul Zolkind, Ann Marie Egloff, Katie M. Campbell, and Ravindra Uppaluri
- Abstract
Supplementary Figure
- Published
- 2023
63. Supplementary Figure 8 from Neoadjuvant and Adjuvant Pembrolizumab in Resectable Locally Advanced, Human Papillomavirus–Unrelated Head and Neck Cancer: A Multicenter, Phase II Trial
- Author
-
Douglas R. Adkins, Obi L. Griffith, Malachi Griffith, Rebecca D. Chernock, Jay F. Piccirillo, Dorina Kallogjeri, Ian S. Hagemann, Scott J. Rodig, Mackenzie Daly, Hiram A. Gay, Wade Thorstad, Ana Lako, Evisa Gjini, Jonathan D. Schoenfeld, Robert Haddad, Jason Kass, Glenn J. Hanna, Matthew D. Stachler, Vickie Y. Jo, Liye Zhou, Rachel Riley, Tenny Mudianto, Trevor J. Pugh, Iulia Cirlan, Youstina Hanna, David T. Mulder, Tiantian Li, Tianxiang Lin, Nicholas C. Spies, Erica K. Barnell, Gavin P. Dunn, Peter Oppelt, Jessica Ley, Loren S. Michel, Patrik Pipkorn, Ryan Jackson, Jason T. Rich, Randal C. Paniello, Brian Nussenbaum, Zachary L. Skidmore, Paul Zolkind, Ann Marie Egloff, Katie M. Campbell, and Ravindra Uppaluri
- Abstract
Supplementary Figure
- Published
- 2023
64. Supplementary Figure 6 from T-Cell Exhaustion Signatures Vary with Tumor Type and Are Severe in Glioblastoma
- Author
-
Peter E. Fecci, Gavin P. Dunn, Kent J. Weinhold, Glenn Dranoff, Patrick Healy, Amber Giles, Darell D. Bigner, Yen-Rei Andrea Yu, Vidyalakshmi Chandramohan, Luis Sanchez-Perez, Tanner M. Johanns, Landon J. Hansen, Christina Jackson, Shohei Koyama, Xiuyu Cui, Aladine A. Elsamadicy, S. Harrison Farber, Cosette Dechant, Hanna Kemeny, Kristen Rhodin, Pakawat Chongsathidkiet, and Karolina Woroniecka
- Abstract
T cell exhaustion in brain metastases models
- Published
- 2023
65. Supplementary Table 1 from T-Cell Exhaustion Signatures Vary with Tumor Type and Are Severe in Glioblastoma
- Author
-
Peter E. Fecci, Gavin P. Dunn, Kent J. Weinhold, Glenn Dranoff, Patrick Healy, Amber Giles, Darell D. Bigner, Yen-Rei Andrea Yu, Vidyalakshmi Chandramohan, Luis Sanchez-Perez, Tanner M. Johanns, Landon J. Hansen, Christina Jackson, Shohei Koyama, Xiuyu Cui, Aladine A. Elsamadicy, S. Harrison Farber, Cosette Dechant, Hanna Kemeny, Kristen Rhodin, Pakawat Chongsathidkiet, and Karolina Woroniecka
- Abstract
Patient and Control Characteristics
- Published
- 2023
66. Supplementary Methods, Figures 1-10 from ERK1/2 Regulation of CD44 Modulates Oral Cancer Aggressiveness
- Author
-
Ravindra Uppaluri, John B. Sunwoo, Jack D. Bui, Gavin P. Dunn, James S. Lewis, Jonathan H. Law, Joshua J. Brotman, Oihana Murillo-Sauca, Ashley E. Winkler, and Nancy P. Judd
- Abstract
PDF file - 2.3MB
- Published
- 2023
67. Integrated analysis of genomic and transcriptomic data for the discovery of splice-associated variants in cancer
- Author
-
Kelsy C. Cotto, Yang-Yang Feng, Avinash Ramu, Megan Richters, Sharon L. Freshour, Zachary L. Skidmore, Huiming Xia, Joshua F. McMichael, Jason Kunisaki, Katie M. Campbell, Timothy Hung-Po Chen, Emily B. Rozycki, Douglas Adkins, Siddhartha Devarakonda, Sumithra Sankararaman, Yiing Lin, William C. Chapman, Christopher A. Maher, Vivek Arora, Gavin P. Dunn, Ravindra Uppaluri, Ramaswamy Govindan, Obi L. Griffith, and Malachi Griffith
- Subjects
Multidisciplinary ,General Physics and Astronomy ,General Chemistry ,General Biochemistry, Genetics and Molecular Biology - Abstract
Somatic mutations within non-coding regions and even exons may have unidentified regulatory consequences that are often overlooked in analysis workflows. Here we present RegTools (www.regtools.org), a computationally efficient, free, and open-source software package designed to integrate somatic variants from genomic data with splice junctions from bulk or single cell transcriptomic data to identify variants that may cause aberrant splicing. We apply RegTools to over 9000 tumor samples with both tumor DNA and RNA sequence data. RegTools discovers 235,778 events where a splice-associated variant significantly increases the splicing of a particular junction, across 158,200 unique variants and 131,212 unique junctions. To characterize these somatic variants and their associated splice isoforms, we annotate them with the Variant Effect Predictor, SpliceAI, and Genotype-Tissue Expression junction counts and compare our results to other tools that integrate genomic and transcriptomic data. While many events are corroborated by the aforementioned tools, the flexibility of RegTools also allows us to identify splice-associated variants in known cancer drivers, such as TP53, CDKN2A, and B2M, and other genes.
- Published
- 2023
68. A Low Subfrontal Dural Opening for Operative Management of Anterior Skull Base Lesions
- Author
-
Samuel J. Cler, Gavin P. Dunn, Gregory J. Zipfel, Ralph G. Dacey, and Michael R. Chicoine
- Subjects
Neurology (clinical) - Abstract
Introduction A low subfrontal dural opening technique that limits brain manipulation was assessed in patients who underwent frontotemporal approaches for anterior fossa lesions. Methods A retrospective review was performed for cases using a low subfrontal dural opening including characterization of demographics, lesion size and location, neurological and ophthalmological assessments, clinical course, and imaging findings. Results A low subfrontal dural opening was performed in 23 patients (17F, 6M), median age of 53 years (range 23–81) with a median follow-up duration of 21.9 months (range 6.2–67.1). Lesions included 22 meningiomas (nine anterior clinoid, 12 tuberculum sellae, and one sphenoid wing), one unruptured internal carotid artery aneurysm clipped during a meningioma resection, and one optic nerve cavernous malformation. Maximal possible resection was achieved in all cases including gross total resection in 16/22 (72.7%), near total in 1/22 (4.5%), and subtotal in 5/22 (22.7%) in which tumor involvement of critical structures limited complete resection. Eighteen patients presented with vision loss; 11 (61%) improved postoperatively, three (17%) were stable, and four (22%) worsened. The mean ICU stay and time to discharge were 1.3 days (range 0–3) and 3.8 days (range 2–8). Conclusion A low sub-frontal dural opening for approaches to the anterior fossa can be performed with minimal brain exposure, early visualization of the optico-carotid cistern for cerebrospinal fluid release, minimizing need for fixed brain retraction, and Sylvian fissure dissection. This technique can potentially reduce surgical risk and provide excellent exposure for anterior skull base lesions with favorable extent of resection, visual recovery, and complication rates.
- Published
- 2022
69. Is There a Role for Immunotherapy in Central Nervous System Cancers?
- Author
-
Gavin P. Dunn, Michael Lim, David A. Reardon, Duane A. Mitchell, Catherine Flores, and Peter E. Fecci
- Subjects
Central Nervous System ,Brain Neoplasms ,business.industry ,medicine.medical_treatment ,Central nervous system ,Immunosuppression ,Hematology ,Newly diagnosed ,Immunotherapy ,medicine.disease ,Cancer Vaccines ,Immune checkpoint ,Central Nervous System Neoplasms ,Immune system ,medicine.anatomical_structure ,Oncology ,Negative phase ,medicine ,Cancer research ,Humans ,Neoplasm Recurrence, Local ,Glioblastoma ,business - Abstract
Glioblastoma has emerged as an immunotherapy-refractory tumor based on negative phase III studies of anti-programmed cell death-1 therapy among newly diagnosed as well as recurrent patients. In addition, although much work on vaccine and cellular approaches is ongoing, therapeutic benefit with these approaches has been underwhelming. Much scientific insight into the multitiered layers of immunosuppression exploited by glioblastoma tumors is emerging that sheds light on the explanation for the disappointing results to date and highlights possible therapeutic avenues that may offer a better likelihood of therapeutic benefit for immune-based therapies.
- Published
- 2022
70. Therapeutic applications of the cancer immunoediting hypothesis
- Author
-
Gavin P. Dunn, Andrew T. Coxon, and Rupen Desai
- Subjects
0301 basic medicine ,Cancer Research ,Adoptive cell transfer ,medicine.medical_treatment ,Immunologic Surveillance ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Neoplasms ,Tumor Microenvironment ,medicine ,Animals ,Humans ,business.industry ,Cancer ,Immunotherapy ,medicine.disease ,Vaccine therapy ,030104 developmental biology ,Immunoediting ,Immune System ,030220 oncology & carcinogenesis ,Disease Susceptibility ,business ,Carcinogenesis ,Neuroscience - Abstract
Since the late 19th century, the immune system has increasingly garnered interest as a novel avenue for cancer therapy, particularly given scientific breakthroughs in recent decades delineating the fundamental role of the immune system in tumorigenesis. The immunoediting hypothesis has articulated this role, describing three phases of the tumor-immune system interaction: Elimination, Equilibrium, and Escape wherein tumors progress from active immunologic surveillance and destruction through dynamic immunologic stasis to unfettered growth. The primary goals of immunotherapy are to restrict and revert progression through these phases, thereby improving the immune system's ability to control tumor growth. In this review, we detail the development and foundation of the cancer immunoediting hypothesis and apply this hypothesis to the dynamic immunotherapy field that includes checkpoint blockade, vaccine therapy, and adoptive cell transfer.
- Published
- 2022
71. Sonobiopsy for minimally invasive, spatiotemporally-controlled, and sensitive detection of glioblastoma-derived circulating tumor DNA
- Author
-
Rupen Desai, Eric C. Leuthardt, Yimei Yue, Lu Xu, Hong Chen, Michael Talcott, Arash Nazeri, Jinyun Yuan, Aadel A. Chaudhuri, Gavin P. Dunn, H. Michael Gach, Christopher Pham Pacia, and Xiaowei Wang
- Subjects
Brain Neoplasms ,Swine ,business.industry ,glioblastoma mutation ,blood-based liquid biopsy ,Liquid Biopsy ,Medicine (miscellaneous) ,blood-brain barrier ,medicine.disease ,droplet digital PCR ,Circulating Tumor DNA ,Mice ,Sonication ,Circulating tumor DNA ,Cell Line, Tumor ,Cancer research ,Animals ,Humans ,Medicine ,Image-guided focused ultrasound ,Glioblastoma ,business ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Research Paper - Abstract
Though surgical biopsies provide direct access to tissue for genomic characterization of brain cancer, they are invasive and pose significant clinical risks. Brain cancer management via blood-based liquid biopsies is a minimally invasive alternative; however, the blood-brain barrier (BBB) restricts the release of brain tumor-derived molecular biomarkers necessary for sensitive diagnosis. Methods: A mouse glioblastoma multiforme (GBM) model was used to demonstrate the capability of focused ultrasound (FUS)-enabled liquid biopsy (sonobiopsy) to improve the diagnostic sensitivity of brain tumor-specific genetic mutations compared with conventional blood-based liquid biopsy. Furthermore, a pig GBM model was developed to characterize the translational implications of sonobiopsy in humans. Magnetic resonance imaging (MRI)-guided FUS sonication was performed in mice and pigs to locally enhance the BBB permeability of the GBM tumor. Contrast-enhanced T1-weighted MR images were acquired to evaluate the BBB permeability change. Blood was collected immediately after FUS sonication. Droplet digital PCR was used to quantify the levels of brain tumor-specific genetic mutations in the circulating tumor DNA (ctDNA). Histological staining was performed to evaluate the potential for off-target tissue damage by sonobiopsy. Results: Sonobiopsy improved the detection sensitivity of EGFRvIII from 7.14% to 64.71% and TERT C228T from 14.29% to 45.83% in the mouse GBM model. It also improved the diagnostic sensitivity of EGFRvIII from 28.57% to 100% and TERT C228T from 42.86% to 71.43% in the porcine GBM model. Conclusion: Sonobiopsy disrupts the BBB at the spatially-targeted brain location, releases tumor-derived DNA into the blood circulation, and enables timely collection of ctDNA. Converging evidence from both mouse and pig GBM models strongly supports the clinical translation of sonobiopsy for the minimally invasive, spatiotemporally-controlled, and sensitive molecular characterization of brain cancer.
- Published
- 2022
72. Spontaneous rupture of an arachnoid cyst in an adult: illustrative case
- Author
-
Lydia A. Leavitt, Pranav Nanda, Anat Stemmer-Rachamimov, Gavin P. Dunn, and Pamela S. Jones
- Subjects
General Medicine - Abstract
BACKGROUND Arachnoid cysts are common intracranial mass lesions frequently discovered as incidental findings on radiographic imaging. It is routine practice to monitor these lesions as a large majority remain stable. Although traumatic cyst rupture is a known risk, it is rare for patients to present with spontaneous rupture. OBSERVATIONS The authors report the case of a 32-year-old patient who required emergent neurosurgical intervention for spontaneous rupture of a left hemispheric arachnoid cyst. LESSONS Patients with ruptured arachnoid cysts can present with vague, nonspecific symptoms that may delay diagnosis. If not diagnosed and treated promptly, arachnoid cyst rupture can progress to a neurosurgical emergency as the subdural collection may cause extensive mass effect and even cerebral herniation.
- Published
- 2023
73. Tandem chimeric antigen receptor (CAR) T cells targeting EGFRvIII and IL-13Rα2 are effective against heterogeneous glioblastoma
- Author
-
Andrea Schmidts, Ambike A Srivastava, Rishab Ramapriyan, Stefanie R Bailey, Amanda A Bouffard, Daniel P Cahill, Bob S Carter, William T Curry, Gavin P Dunn, Matthew J Frigault, Elizabeth R Gerstner, Jack Y Ghannam, Michael C Kann, Rebecca C Larson, Mark B Leick, Brian V Nahed, Leland G Richardson, Irene Scarfò, Jing Sun, Hiroaki Wakimoto, Marcela V Maus, and Bryan D Choi
- Subjects
Oncology ,Surgery ,Neurology (clinical) - Abstract
Background Chimeric antigen receptor (CAR) T cells have achieved remarkable responses in patients with hematological malignancies; however, the potential of this therapeutic platform for solid tumors like glioblastoma (GBM) has been limited, due in large part to the targeting of single antigens in a heterogeneous disease. Strategies that allow CAR T cells to engage multiple antigens concomitantly may broaden therapeutic responses and mitigate the effects of immune escape. Methods Here we have developed a novel, dual-specific, tandem CAR T (TanCART) cell with the ability to simultaneously target both EGFRvIII and IL-13Rα2, two well-characterized tumor antigens that are frequently found on the surface of GBM cells but completely absent from normal brain tissues. We employed both standard immunological assays and multiple orthotopic preclinical models including patient-derived xenograft to demonstrate efficacy of this approach against heterogeneous tumors. Results Tandem CAR T cells displayed enhanced cytotoxicity in vitro against heterogeneous GBM populations, including patient-derived brain tumor cultures (P < .05). Compared to CAR T cells targeting single antigens, dual antigen engagement through the tandem construct was necessary to achieve long-term, complete, and durable responses in orthotopic murine models of heterogeneous GBM, including patient-derived xenografts (P < .05). Conclusions We demonstrate that TanCART is effective against heterogeneous tumors in the brain. These data lend further credence to the development of multi-specific CAR T cells in the treatment of GBM and other cancers.
- Published
- 2022
74. 463 Tandem CAR T Cells Targeting EGFRvIII and IL-13R⍺2 are Effective Against Heterogeneous Glioblastoma
- Author
-
Andrea Schmidts, Ambike Srivastava, Rishab Ramapriyan, Daniel P. Cahill, Bob S. Carter, William T. Curry, Gavin P. Dunn, Jack Ghannam, Brian V. Nahed, Jing Sun, Hiroaki Wakimoto, Marcela V. Maus, and Bryan D. Choi
- Subjects
Surgery ,Neurology (clinical) - Published
- 2023
75. Characterization of the Genomic and Immunologic Diversity of Malignant Brain Tumors through Multisector Analysis
- Author
-
Anthony Z. Wang, Hsiang Chih Lu, Zachary L. Skidmore, Obi L. Griffith, Gregory J. Zipfel, Katherine E. Miller, Megan Richters, Eric C. Leuthardt, Malachi Griffith, Albert H. Kim, Gavin P. Dunn, Tanner M. Johanns, Tammi L. Vickery, Joshua W. Osbun, Ralph G. Dacey, Bryan Fisk, Elaine R. Mardis, Michael R. Chicoine, Joshua L. Dowling, and Maximilian Schaettler
- Subjects
Brain Neoplasms ,Immune microenvironment ,T cell ,Receptors, Antigen, T-Cell ,Genomics ,Biology ,medicine.disease ,Article ,Immunological diversity ,Immune system ,medicine.anatomical_structure ,Oncology ,Exome Sequencing ,Tumor Microenvironment ,Cancer research ,medicine ,Humans ,Immunotherapy ,Neoplasm Metastasis ,Treatment resistance ,Glioblastoma ,Multi sectoral ,Exome - Abstract
Despite some success in secondary brain metastases, targeted or immune-based therapies have shown limited efficacy against primary brain malignancies such as glioblastoma (GBM). Although the intratumoral heterogeneity of GBM is implicated in treatment resistance, it remains unclear whether this diversity is observed within brain metastases and to what extent cancer cell–intrinsic heterogeneity sculpts the local immune microenvironment. Here, we profiled the immunogenomic state of 93 spatially distinct regions from 30 malignant brain tumors through whole-exome, RNA, and T-cell receptor sequencing. Our analyses identified differences between primary and secondary malignancies, with gliomas displaying more spatial heterogeneity at the genomic and neoantigen levels. In addition, this spatial diversity was recapitulated in the distribution of T-cell clones in which some gliomas harbored highly expanded but spatially restricted clonotypes. This study defines the immunogenomic landscape across a cohort of malignant brain tumors and contains implications for the design of targeted and immune-based therapies against intracranial malignancies. Significance: This study describes the impact of spatial heterogeneity on genomic and immunologic characteristics of gliomas and brain metastases. The results suggest that gliomas harbor significantly greater intratumoral heterogeneity of genomic alterations, neoantigens, and T-cell clones than brain metastases, indicating the importance of multisector analysis for clinical or translational studies. This article is highlighted in the In This Issue feature, p. 1
- Published
- 2021
76. Internal dose escalation associated with increased local control for melanoma brain metastases treated with stereotactic radiosurgery
- Author
-
Gavin P. Dunn, S. Murty Goddu, Joshua L. Dowling, Clifford G. Robinson, Timothy J. Mitchell, J. Kavanaugh, Todd A. DeWees, Mustafaa Mahmood, Keith M. Rich, Michael R. Chicoine, Eric C. Leuthardt, Albert H. Kim, Christopher Abraham, Christina Tsien, Stephanie M. Perkins, William R. Kennedy, Jiayi Huang, Sahaja Acharya, and Nels C. Knutson
- Subjects
medicine.medical_specialty ,Proportional hazards model ,business.industry ,Melanoma ,medicine.medical_treatment ,Urology ,General Medicine ,medicine.disease ,Radiosurgery ,Targeted therapy ,Lesion ,03 medical and health sciences ,0302 clinical medicine ,Unresected ,030220 oncology & carcinogenesis ,Toxicity ,medicine ,medicine.symptom ,Radiation treatment planning ,business ,030217 neurology & neurosurgery - Abstract
OBJECTIVE The internal high-dose volume varies widely for a given prescribed dose during stereotactic radiosurgery (SRS) to treat brain metastases (BMs). This may be altered during treatment planning, and the authors have previously shown that this improves local control (LC) for non–small cell lung cancer BMs without increasing toxicity. Here, they seek to identify potentially actionable dosimetric predictors of LC after SRS for melanoma BM. METHODS The records of patients with unresected melanoma BM treated with single-fraction Gamma Knife RS between 2006 and 2017 were reviewed. LC was assessed on a per-lesion basis, defined as stability or a decrease in lesion size. Outcome-oriented approaches were utilized to determine optimal dichotomization for dosimetric variables relative to LC. Univariable and multivariable Cox regression analysis was implemented to evaluate the impact of collected parameters on LC. RESULTS Two hundred eighty-seven melanoma BMs in 79 patients were identified. The median age was 56 years (range 31–86 years). The median follow-up was 7.6 months (range 0.5–81.6 months), and the median survival was 9.3 months (range 1.3–81.6 months). Lesions were optimally stratified by volume receiving at least 30 Gy (V30) greater than or equal to versus less than 25%. V30 was ≥ and < 25% in 147 and 140 lesions, respectively. For all patients, 1-year LC was 83% versus 66% for V30 ≥ and < 25%, respectively (p = 0.001). Stratifying by volume, lesions 2 cm or less (n = 215) had 1-year LC of 82% versus 70% (p = 0.013) for V30 ≥ and < 25%, respectively. Lesions > 2 to 3 cm (n = 32) had 1-year LC of 100% versus 43% (p = 0.214) for V30 ≥ and < 25%, respectively. V30 was still predictive of LC even after controlling for the use of immunotherapy and targeted therapy. Radionecrosis occurred in 2.8% of lesions and was not significantly associated with V30. CONCLUSIONS For a given prescription dose, an increased internal high-dose volume, as indicated by measures such as V30 ≥ 25%, is associated with improved LC but not increased toxicity in single-fraction SRS for melanoma BM. Internal dose escalation is an independent predictor of improved LC even in patients receiving immunotherapy and/or targeted therapy. This represents a dosimetric parameter that is actionable at the time of treatment planning and warrants further evaluation.
- Published
- 2021
77. Editorial: It takes a village: The expanding multi-disciplinary approach to brain metastasis
- Author
-
Peter E, Fecci, Ganesh, Rao, Priscilla K, Brastianos, Gavin P, Dunn, and Carey K, Anders
- Subjects
Cancer Research ,Oncology - Published
- 2022
78. Survival After Resection of Newly-Diagnosed Intracranial Grade II Ependymomas: An Initial Multicenter Analysis and the Logistics of Intraoperative Magnetic Resonance Imaging
- Author
-
Alexander T. Yahanda, Keith M. Rich, Ralph G. Dacey, Gregory J. Zipfel, Gavin P. Dunn, Joshua L. Dowling, Matthew D. Smyth, Eric C. Leuthardt, David D. Limbrick, John Honeycutt, Garnette R. Sutherland, Randy L. Jensen, John Evans, and Michael R. Chicoine
- Subjects
Surgery ,Neurology (clinical) - Abstract
To identify factors, including the use of intraoperative magnetic resonance imaging (iMRI), impacting overall survival (OS) and progression-free survival (PFS) after resections of newly diagnosed intracranial grade II ependymomas performed across 4 different institutions.Analyses of a multicenter mixed retrospective/prospective database assessed the impact of patient, treatment, and tumor characteristics on OS and PFS. iMRI workflow and logistics were also outlined.Forty-three patients were identified (mean age 25.4 years, mean follow-up 52.8 months). The mean OS was 52.8 ± 44.7 months. Univariate analyses failed to identify prognostic factors associated with OS, likely due to relatively shorter follow-up time for this less aggressive glioma subtype. The mean PFS was 43.7 ± 39.8 months. Multivariate analyses demonstrated that gross-total resection was associated with prolonged PFS compared to both subtotal resection (STR) (P = 0.005) and near-total resection (P = 0.01). Infratentorial location was associated with improved PFS compared to supratentorial location (P = 0.04). Log-rank analyses of Kaplan-Meier survival curves showed that increasing extent of resection (EOR) led to improved OS specifically for supratentorial tumors (P = 0.02) and improved PFS for all tumors (P0.001). Thirty cases (69.8%) utilized iMRI, of which 12 (27.9%) involved additional resection after iMRI. Of these, 8/12 (66.7%) resulted in gross-total resection, while 2/12 (16.7%) were near-total resection and 2/12 (16.7%) were subtotal resection. iMRI was not an independent prognosticator of PFS (P = 0.72).Greater EOR and infratentorial location were associated with increased PFS for grade II ependymomas. Greater EOR was associated with longer OS only for supratentorial tumors. A longer follow-up is needed to establish prognostic factors for this cohort, including use of iMRI.
- Published
- 2022
79. Re-evaluating Biopsy for Recurrent Glioblastoma: A Position Statement by the Christopher Davidson Forum Investigators
- Author
-
Constantinos G. Hadjipanayis, Analiz Rodriguez, Jian Campian, Melanie Hayden Gephart, Daniel A. Orringer, Jennifer S. Yu, Ali Jalali, James Battiste, Gavin P. Dunn, Akash J. Patel, Peter E. Fecci, Dimitris G. Placantonakis, Eric C. Leuthardt, Albert H. Kim, Sunit Das, Kimberly B Hoang, Mario L. Suvà, Ralph G. Dacey, Milan G. Chheda, Greg Zipfel, Nduka Amankulor, Isaac Yang, and Edjah K. Nduom
- Subjects
Position statement ,medicine.medical_specialty ,Stereotactic biopsy ,Biopsy ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Intensive care medicine ,Pseudoprogression ,medicine.diagnostic_test ,Brain Neoplasms ,business.industry ,Recurrent glioblastoma ,medicine.disease ,Clinical trial ,030220 oncology & carcinogenesis ,Mutation ,Surgery ,Neurology (clinical) ,Personalized medicine ,Neoplasm Recurrence, Local ,Glioblastoma ,business ,030217 neurology & neurosurgery - Abstract
Patients with glioblastoma (GBM) need bold new approaches to their treatment, yet progress has been hindered by a relative inability to dynamically track treatment response, mechanisms of resistance, evolution of targetable mutations, and changes in mutational burden. We are writing on behalf of a multidisciplinary group of academic neuro-oncology professionals who met at the collaborative Christopher Davidson Forum at Washington University in St Louis in the fall of 2019. We propose a dramatic but necessary change to the routine management of patients with GBM to advance the field: to routinely biopsy recurrent GBM at the time of presumed recurrence. Data derived from these samples will identify true recurrence vs treatment effect, avoid treatments with little chance of success, enable clinical trial access, and aid in the scientific advancement of our understanding of GBM.
- Published
- 2021
80. A pilot study of lymphoscintigraphy with tracer injection into the human brain
- Author
-
Andrew T Coxon, Rupen Desai, Pujan R Patel, Ananth K Vellimana, Jon T Willie, Joshua L Dowling, Eric C Leuthardt, Albert H Kim, Tanner M Johanns, Barry A Siegel, and Gavin P Dunn
- Subjects
Neurology ,Neurology (clinical) ,Cardiology and Cardiovascular Medicine - Abstract
Many groups have reported lymphatic and glymphatic structures in animal and human brains, but tracer injection into the human brain to demonstrate real-time lymphatic drainage and mapping has not been described. We enrolled patients undergoing standard-of-care resection or stereotactic biopsy for suspected intracranial tumors. Patients received peritumoral injections of 99mTc-tilmanocept followed by planar or tomographic imaging. Fourteen patients with suspected brain tumors were enrolled. One was excluded from analysis because of tracer leakage during injection. There was no drainage of 99mTc-tilmanocept to regional lymph nodes in any of the patients. On average, after correcting for radioactive decay, 70.7% (95% CI: 59.9%, 81.6%) of the tracer in the injection site and 78.1% (95% CI: 71.1%, 85.1%) in the whole-head on the day of surgery remained the morning after, with variable radioactivity in the subarachnoid space. The retained fraction was much greater than expected based on the clearance rate from non-brain injection sites. In this pilot study, the lymphatic tracer 99mTc-tilmanocept was injected into the brain parenchyma, and there was no drainage outside the brain to the cervical lymph nodes. Our work demonstrates an inefficiency of drainage from peritumoral brain parenchyma and highlights a therapeutic opportunity to improve immunosurveillance of the brain.
- Published
- 2023
81. Yap1 Mediates Trametinib Resistance in Head and Neck Squamous Cell Carcinomas
- Author
-
Paul Zolkind, Gavin P. Dunn, Ann Marie Egloff, Erica K. Barnell, Ravindra Uppaluri, Obi L. Griffith, Tenny Mudianto, Tusar Giri, Rachel S. Riley, Malachi Griffith, Jason Webb, Zachary L. Skidmore, Douglas Adkins, Katie M. Campbell, and Ibrahim Ozgenc
- Subjects
0301 basic medicine ,Cancer Research ,MAP Kinase Signaling System ,Pyridones ,Biopsy ,Cell ,Pyrimidinones ,Article ,Gene Knockout Techniques ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Exome Sequencing ,Animals ,Humans ,Medicine ,Hippo Signaling Pathway ,RNA-Seq ,Trametinib ,YAP1 ,Hippo signaling pathway ,Squamous Cell Carcinoma of Head and Neck ,business.industry ,YAP-Signaling Proteins ,medicine.disease ,Xenograft Model Antitumor Assays ,Head and neck squamous-cell carcinoma ,Verteporfin ,Gene Expression Regulation, Neoplastic ,Clinical trial ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Drug Resistance, Neoplasm ,Head and Neck Neoplasms ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,Female ,business ,medicine.drug - Abstract
Purpose: In a head and neck squamous cell carcinoma (HNSCC) “window of opportunity” clinical trial, we reported that trametinib reduced MEK-Erk1/2 activation and resulted in tumor responses in a subset of patients. Here, we investigated resistance to trametinib and molecular correlates in HNSCC cell lines and patient samples. Experimental Design: HNSCC cell lines were treated with trametinib to generate resistant lines. Candidate bypass pathways were assessed using immunoblotting, CRISPR knockout, and survival assays. Effectiveness of combined trametinib and verteporfin targeting was evaluated. Patient-derived xenografts (PDXs) from responder patients were treated with trametinib and resistant tumors were analyzed. Window trial clinical samples were subjected to whole-exome and RNA sequencing. Results: HNSCC cell lines developed resistance (CAL27-TR and HSC3-TR) after prolonged trametinib exposure. Downstream effectors of the Hippo pathway were activated in CAL27-TR and HSC3-TR, and combined trametinib and verteporfin treatment resulted in synergistic treatment response. We defined the Hippo pathway effector Yap1 as an induced survival pathway promoting resistance to trametinib in HSC3-TR. Yap1 was necessary for HSC3-TR trametinib resistance, and constitutively active Yap1 was sufficient to confer resistance in parental HSC3. Analysis of trametinib neoadjuvant trial patient tumors indicated canonical MEK-Erk1/2 pathway activating mutations were infrequent, and Yap1 activity increased following trametinib treatment. Trametinib treatment of a PDX from a responder patient resulted in evolution of resistance with increased Yap1 expression and activity. Conclusions: These studies identify a Yap1-dependent resistance to trametinib therapy in HNSCCs. Combined Yap1 and MEK targeting may represent a strategy to enhance HNSCC response.
- Published
- 2021
82. Immune profiling of pituitary tumors reveals variations in immune infiltration and checkpoint molecule expression
- Author
-
Wenya Linda Bi, Yu Mei, David Meredith, James Agolia, Malak Abedalthagafi, Changchen Hu, Alexandra M Giantini Larsen, Sally Al Abdulmohsen, Gavin P. Dunn, Tejus Bale, and Ian F. Dunn
- Subjects
LAG3 ,business.industry ,Angiogenesis ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,T cell ,Pituitary tumors ,030209 endocrinology & metabolism ,Immunotherapy ,medicine.disease ,Immune checkpoint ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Immune system ,medicine.anatomical_structure ,Pituitary adenoma ,medicine ,Cancer research ,business ,030217 neurology & neurosurgery - Abstract
Pituitary tumors are the second most common primary brain tumors. Functional tumors demonstrate increased PD-L1 expression, but expression of other checkpoint regulators has not been characterized. We sought to characterize the immune microenvironment of human pituitary tumors to identify new treatment opportunities. 72 pituitary tumors were evaluated for expression of the immune regulatory markers programmed death ligand 1 (PD-L1), programmed death ligand 2 (PD-L2), V-domain Ig suppressor of T cell activation (VISTA), lymphocyte activation gene 3 (LAG3) and tumor necrosis factor receptor superfamily member 4 (OX40) by immunohistochemistry (IHC). Lymphocyte infiltration, macrophage infiltration, and angiogenesis were analyzed using IHC. Expression of pituitary tumor initiating cell marker CD15 and mismatch repair proteins MutS protein homolog 2 (MSH2) and MutS protein homolog 6 (MSH6) was also assessed. Pituitary tumors were infiltrated by macrophages and T cells, and they expressed varying levels of PD-L1, PD-L2, VISTA, LAG3, and OX40. Functional tumors and tumors with high expression of tumor stem cell markers had higher immune cell infiltration and greater expression of immunosuppressive checkpoint regulators. Increased PD-L1 and LAG3 and reduced VISTA were observed in primary tumors compared to recurrent tumors. Immune cell infiltration and checkpoint regulator expression vary depending on functional status and presence of pituitary tumor initiating cells. Functional tumors may have a particularly immunosuppressive microenvironment. Further studies of immune checkpoint blockade of pituitary tumors, particularly functional tumors, are warranted, though combination therapy may be required.
- Published
- 2021
83. Cancers from Novel Pole-Mutant Mouse Models Provide Insights into Polymerase-Mediated Hypermutagenesis and Immune Checkpoint Blockade
- Author
-
Jonathan N. Byrd, James G. Jackson, Cynthia Hawkins, Alberto Martin, Uri Tabori, Walter J. Zahurancik, Dana Elshaer, Cynthia J. Guidos, Melissa Galati, Sumedha Sudhaman, Oz Mordechai, Ayse Bahar Ercan, Tomasz Sarosiek, Melyssa Aronson, Miki S. Gams, Victoria J. Forster, Taylor Bridge, Eric Bouffet, Andrea H. Seeley, Carol Durno, Melissa Edwards, Nathan Ungerleider, Vivian S. Park, Adam Shlien, Zachary F. Pursell, Lazar Joksimovic, Robert Siddaway, Nuno Miguel Nunes, Xia Wang, Zucai Suo, Iram Siddiqui, Jagadeesh Ramdas, Gavin P. Dunn, Jiil Chung, Richard de Borja, and Karl P. Hodel
- Subjects
0301 basic medicine ,Cancer Research ,Mechanism (biology) ,medicine.medical_treatment ,Mutant ,Mutagenesis (molecular biology technique) ,Cancer ,Immunotherapy ,Biology ,medicine.disease_cause ,medicine.disease ,Immune checkpoint ,3. Good health ,Blockade ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,Carcinogenesis - Abstract
POLE mutations are a major cause of hypermutant cancers, yet questions remain regarding mechanisms of tumorigenesis, genotype–phenotype correlation, and therapeutic considerations. In this study, we establish mouse models harboring cancer-associated POLE mutations P286R and S459F, which cause rapid albeit distinct time to cancer initiation in vivo, independent of their exonuclease activity. Mouse and human correlates enabled novel stratification of POLE mutations into three groups based on clinical phenotype and mutagenicity. Cancers driven by these mutations displayed striking resemblance to the human ultrahypermutation and specific signatures. Furthermore, Pole-driven cancers exhibited a continuous and stochastic mutagenesis mechanism, resulting in intertumoral and intratumoral heterogeneity. Checkpoint blockade did not prevent Pole lymphomas, but rather likely promoted lymphomagenesis as observed in humans. These observations provide insights into the carcinogenesis of POLE-driven tumors and valuable information for genetic counseling, surveillance, and immunotherapy for patients. Significance: Two mouse models of polymerase exonuclease deficiency shed light on mechanisms of mutation accumulation and considerations for immunotherapy. See related commentary by Wisdom and Kirsch p. 5459
- Published
- 2020
84. 33. Computational prediction of MHC anchor locations guide neoantigen identification and prioritization
- Author
-
Huiming Xia, Joshua McMichael, Michelle Becker-Hapak, Onyinyechi C. Onyeador, Rico Buchli, Ethan McClain, Patrick Pence, Suangson Supabphol, Megan M. Richters, Anamika Basu, Cody A. Ramirez, Cristina Puig-Saus, Kelsy C. Cotto, Jasreet Hundal, Susanna Kiwala, S. Peter Goedegebuure, Tanner M. Johanns, Gavin P. Dunn, Antoni Ribas, Christopher A. Miller, William E. Gillanders, Todd A. Fehniger, Obi L. Griffith, and Malachi Griffith
- Subjects
Cancer Research ,Genetics ,Molecular Biology - Published
- 2022
85. Intraoperative MRI for newly diagnosed supratentorial glioblastoma: a multicenter-registry comparative study to conventional surgery
- Author
-
John Honeycutt, Michael R. Chicoine, Eric C. Leuthardt, Joshua L. Dowling, Peter T Sylvester, John J. Evans, Albert H. Kim, Gregory J. Zipfel, Daniel P. Cahill, Alexander T. Yahanda, Robert L. Grubb, Steven R Abram, Amar S Shah, Mark C. Oswood, Mitesh V. Shah, Ralph G. Dacey, Garnette R. Sutherland, Gavin P. Dunn, Keith M. Rich, Yu Tao, Ananth K. Vellimana, and Randy L. Jensen
- Subjects
medicine.medical_specialty ,IDH1 ,Multivariate analysis ,business.industry ,Proportional hazards model ,Interventional magnetic resonance imaging ,Retrospective cohort study ,General Medicine ,Intraoperative MRI ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Medicine ,Radiology ,Neurosurgery ,business ,Supratentorial Glioblastoma ,030217 neurology & neurosurgery - Abstract
OBJECTIVEIntraoperative MRI (iMRI) is used in the surgical treatment of glioblastoma, with uncertain effects on outcomes. The authors evaluated the impact of iMRI on extent of resection (EOR) and overall survival (OS) while controlling for other known and suspected predictors.METHODSA multicenter retrospective cohort of 640 adult patients with newly diagnosed supratentorial glioblastoma who underwent resection was evaluated. iMRI was performed in 332/640 cases (51.9%). Reviews of MRI features and tumor volumetric analysis were performed on a subsample of cases (n = 286; 110 non-iMRI, 176 iMRI) from a single institution.RESULTSThe median age was 60.0 years (mean 58.5 years, range 20.5–86.3 years). The median OS was 17.0 months (95% CI 15.6–18.4 months). Gross-total resection (GTR) was achieved in 403/640 cases (63.0%). Kaplan-Meier analysis of 286 cases with volumetric analysis for EOR (grouped into 100%, 95%–99%, 80%–94%, and 50%–79%) showed longer OS for 100% EOR compared to all other groups (p < 0.01). Additional resection after iMRI was performed in 104/122 cases (85.2%) with initial subtotal resection (STR), leading to a 6.3% mean increase in EOR and a 2.2-cm3 mean decrease in tumor volume. For iMRI cases with volumetric analysis, the GTR rate increased from 54/176 (30.7%) on iMRI to 126/176 (71.5%) postoperatively. The EOR was significantly higher in the iMRI group for intended GTR and STR groups (p = 0.02 and p < 0.01, respectively). Predictors of GTR on multivariate logistic regression included iMRI use and intended GTR. Predictors of shorter OS on multivariate Cox regression included older age, STR, isocitrate dehydrogenase 1 (IDH1) wild type, no O6-methylguanine DNA methyltransferase (MGMT) methylation, and no Stupp therapy. iMRI was a significant predictor of OS on univariate (HR 0.82, 95% CI 0.69–0.98; p = 0.03) but not multivariate analyses. Use of iMRI was not associated with an increased rate of new permanent neurological deficits.CONCLUSIONSGTR increased OS for patients with newly diagnosed glioblastoma after adjusting for other prognostic factors. iMRI increased EOR and GTR rate and was a significant predictor of GTR on multivariate analysis; however, iMRI was not an independent predictor of OS. Additional supporting evidence is needed to determine the clinical benefit of iMRI in the management of glioblastoma.
- Published
- 2020
86. Impact of Intraoperative Magnetic Resonance Imaging and Other Factors on Surgical Outcomes for Newly Diagnosed Grade II Astrocytomas and Oligodendrogliomas: A Multicenter Study
- Author
-
Gregory J. Zipfel, John Honeycutt, Matthew D. Smyth, Mitesh V. Shah, Randy L. Jensen, Bhuvic Patel, Eric C. Leuthardt, Keith M. Rich, Albert H. Kim, David D. Limbrick, Steven R Abram, Daniel P. Cahill, Joshua L. Dowling, Alexander T. Yahanda, Amar S Shah, Michael R. Chicoine, John J. Evans, Garnette R. Sutherland, Jeffrey R. Leonard, Ralph G. Dacey, and Gavin P. Dunn
- Subjects
Adult ,medicine.medical_specialty ,Adolescent ,Neuroimaging ,Kaplan-Meier Estimate ,Gastroenterology ,Neurosurgical Procedures ,Intraoperative MRI ,Young Adult ,Glioma ,Internal medicine ,medicine ,Grade II Glioma ,Humans ,Child ,neoplasms ,Aged ,Retrospective Studies ,medicine.diagnostic_test ,Brain Neoplasms ,business.industry ,Hazard ratio ,Astrocytoma ,Magnetic resonance imaging ,Odds ratio ,Middle Aged ,medicine.disease ,Magnetic Resonance Imaging ,Progression-Free Survival ,Surgery, Computer-Assisted ,Child, Preschool ,Female ,Surgery ,Neurology (clinical) ,Oligodendroglioma ,business - Abstract
BACKGROUND Few studies use large, multi-institutional patient cohorts to examine the role of intraoperative magnetic resonance imaging (iMRI) in the resection of grade II gliomas. OBJECTIVE To assess the impact of iMRI and other factors on overall survival (OS) and progression-free survival (PFS) for newly diagnosed grade II astrocytomas and oligodendrogliomas. METHODS Retrospective analyses of a multicenter database assessed the impact of patient-, treatment-, and tumor-related factors on OS and PFS. RESULTS A total of 232 resections (112 astrocytomas and 120 oligodendrogliomas) were analyzed. Oligodendrogliomas had longer OS (P
- Published
- 2020
87. Emerging immunotherapies for malignant glioma: from immunogenomics to cell therapy
- Author
-
David A. Reardon, Timothy F. Cloughesy, Gavin P. Dunn, Robert M. Prins, Adam M. Sonabend, and Marcela V. Maus
- Subjects
Cancer Research ,business.industry ,T cell ,medicine.medical_treatment ,Cell- and Tissue-Based Therapy ,Cancer ,Review ,Glioma ,Immunotherapy ,Bioinformatics ,medicine.disease ,Chimeric antigen receptor ,Immune checkpoint ,Cell therapy ,medicine.anatomical_structure ,Immune system ,Oncology ,medicine ,Humans ,Neurology (clinical) ,Glioblastoma ,business - Abstract
As immunotherapy assumes a central role in the management of many cancers, ongoing work is directed at understanding whether immune-based treatments will be successful in patients with glioblastoma (GBM). Despite several large studies conducted in the last several years, there remain no FDA-approved immunotherapies in this patient population. Nevertheless, there are a range of exciting new approaches being applied to GBM, all of which may not only allow us to develop new treatments but also help us understand fundamental features of the immune response in the central nervous system. In this review, we summarize new developments in the application of immune checkpoint blockade, from biomarker-driven patient selection to the timing of treatment. Moreover, we summarize novel work in personalized immune-oncology by reviewing work in cancer immunogenomics–driven neoantigen vaccine studies. Finally, we discuss cell therapy efforts by reviewing the current state of chimeric antigen receptor T-cell therapy.
- Published
- 2020
88. A review of glioblastoma immunotherapy
- Author
-
Maryam Rahman, Peter E. Fecci, Ravi Medikonda, Michael Lim, and Gavin P. Dunn
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,Combination therapy ,medicine.medical_treatment ,03 medical and health sciences ,0302 clinical medicine ,Renal cell carcinoma ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,medicine ,Humans ,Brain Neoplasms ,business.industry ,Melanoma ,Immunotherapy ,medicine.disease ,Vaccine therapy ,Immune checkpoint ,Radiation therapy ,Clinical trial ,Neurology ,030220 oncology & carcinogenesis ,Neurology (clinical) ,Neoplasm Recurrence, Local ,Glioblastoma ,business ,030217 neurology & neurosurgery - Abstract
Glioblastoma is a very aggressive cancer with dismal prognosis despite standard of care including surgical resection, radiation therapy, and chemotherapy. There is interest in applying immunotherapy to glioblastoma as this modality has demonstrated remarkable improvements in the management of several solid tumors including melanoma, renal cell carcinoma, and non-small cell lung cancer. This review aims to provide an overview of the current state of glioblastoma immunotherapy. Literature search was performed on PubMed between 1961 and 2020. Initial clinical trials of checkpoint inhibitors and vaccine therapy for glioblastoma have largely been disappointing for both primary and recurrent glioblastoma. This failure has been attributed to glioblastoma’s highly immunosuppressive environment and multiple mechanisms of therapy resistance including high tumor heterogeneity, low mutational burden, systemic immunosuppression, and local immune dysfunction. Current clinical trials are exploring combination therapy and novel treatment strategies beyond immune checkpoint therapies and vaccine therapy such as CAR T cells. There is also an effort to establish synergy between immunotherapy and current standard of care. Furthermore, recent advances in personalized neoantigen vaccines suggest a shift towards personalized, patient-specific GBM treatment.
- Published
- 2020
89. Consensus recommendations for a standardized brain tumor imaging protocol for clinical trials in brain metastases
- Author
-
Christina Tsien, Jerrold L. Boxerman, Evanthia Galanis, Terry C. Burns, Nan Lin, Caroline Chung, Michael Weller, Patrick Y. Wen, Benjamin M. Ellingson, Daniel P. Barboriak, Paul D. Brown, Ian F. Parney, Elizabeth R. Gerstner, Lalitha K. Shankar, Timothy J. Kaufmann, Raymond Y. Huang, Martin J. van den Bent, Marion Smits, Gavin P. Dunn, Priscilla K. Brastianos, Radiology & Nuclear Medicine, and Neurology
- Subjects
Cancer Research ,medicine.medical_specialty ,Consensus ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Brain tumor ,Reviews ,Contrast Media ,Gadolinium ,Fluid-attenuated inversion recovery ,Tumor response ,03 medical and health sciences ,0302 clinical medicine ,Rare Diseases ,Clinical Research ,brain metastases ,medicine ,Medical imaging ,Image acquisition ,Humans ,protocol ,Oncology & Carcinogenesis ,Cancer ,Protocol (science) ,medicine.diagnostic_test ,business.industry ,Brain Neoplasms ,Neurosciences ,imaging ,Magnetic resonance imaging ,medicine.disease ,Magnetic Resonance Imaging ,Brain Disorders ,Clinical trial ,Brain Cancer ,Oncology ,030220 oncology & carcinogenesis ,Biomedical Imaging ,Neurology (clinical) ,Radiology ,business ,030217 neurology & neurosurgery ,MRI - Abstract
A recent meeting was held on March 22, 2019, among the FDA, clinical scientists, pharmaceutical and biotech companies, clinical trials cooperative groups, and patient advocacy groups to discuss challenges and potential solutions for increasing development of therapeutics for central nervous system metastases. A key issue identified at this meeting was the need for consistent tumor measurement for reliable tumor response assessment, including the first step of standardized image acquisition with an MRI protocol that could be implemented in multicenter studies aimed at testing new therapeutics. This document builds upon previous consensus recommendations for a standardized brain tumor imaging protocol (BTIP) in high-grade gliomas and defines a protocol for brain metastases (BTIP-BM) that addresses unique challenges associated with assessment of CNS metastases. The “minimum standard” recommended pulse sequences include: (i) parameter matched pre- and post-contrast inversion recovery (IR)–prepared, isotropic 3D T1-weighted gradient echo (IR-GRE); (ii) axial 2D T2-weighted turbo spin echo acquired after injection of gadolinium-based contrast agent and before post-contrast 3D T1-weighted images; (iii) axial 2D or 3D T2-weighted fluid attenuated inversion recovery; (iv) axial 2D, 3-directional diffusion-weighted images; and (v) post-contrast 2D T1-weighted spin echo images for increased lesion conspicuity. Recommended sequence parameters are provided for both 1.5T and 3T MR systems. An “ideal” protocol is also provided, which replaces IR-GRE with 3D TSE T1-weighted imaging pre- and post-gadolinium, and is best performed at 3T, for which dynamic susceptibility contrast perfusion is included. Recommended perfusion parameters are given.
- Published
- 2020
90. Glioblastoma Clinical Trials: Current Landscape and Opportunities for Improvement
- Author
-
Rifaquat Rahman, Minesh P. Mehta, David A. Reardon, Louis B. Nabors, Timothy F. Cloughesy, John Sampson, Evanthia Galanis, Susan M. Chang, Roger Stupp, Mustafa Khasraw, Eudocia Q. Lee, Shawn Kothari, Manmeet S Ahluwalia, Stephen J Bagley, Patrick Y. Wen, Stuart A. Grossman, Simon Khagi, Erik P. Sulman, Michael Weller, Gavin P. Dunn, and University of Zurich
- Subjects
Adult ,Cancer Research ,medicine.medical_specialty ,Phase iii trials ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,MEDLINE ,Context (language use) ,610 Medicine & health ,Article ,Adult glioblastoma ,Rare Diseases ,Clinical Research ,Medicine ,Humans ,Medical physics ,Oncology & Carcinogenesis ,Cancer ,business.industry ,Brain Neoplasms ,Neurosciences ,medicine.disease ,Brain Disorders ,10040 Clinic for Neurology ,Clinical trial ,Brain Cancer ,Oncology ,Research Design ,business ,Early phase ,Glioblastoma - Abstract
Therapeutic advances for glioblastoma have been minimal over the past 2 decades. In light of the multitude of recent phase III trials that have failed to meet their primary endpoints following promising preclinical and early-phase programs, a Society for Neuro-Oncology Think Tank was held in November 2020 to prioritize areas for improvement in the conduct of glioblastoma clinical trials. Here, we review the literature, identify challenges related to clinical trial eligibility criteria and trial design in glioblastoma, and provide recommendations from the Think Tank. In addition, we provide a data-driven context with which to frame this discussion by analyzing key study design features of adult glioblastoma clinical trials listed on ClinicalTrials.gov as “recruiting” or “not yet recruiting” as of February 2021.
- Published
- 2022
91. Applied cancer immunogenomics in glioblastoma
- Author
-
Gavin P. Dunn, Connor J. Liu, and Andrew T. Coxon
- Subjects
Tumor microenvironment ,business.industry ,medicine.medical_treatment ,Cancer ,Immunotherapy ,Computational biology ,medicine.disease ,Cell therapy ,Clinical trial ,Immune system ,Antigen ,medicine ,business ,Glioblastoma - Abstract
The use of novel immunotherapeutic agents in treating malignant brain tumors faces considerable challenges. Cancer immunogenomics represents a genomic framework for studying the dynamic interactions between the host immune system and tumor microenvironment. Characterization of the tumor-specific antigen landscape has already yielded a multitude of clinical trials testing personalized glioblastoma vaccines. Furthermore, significant advancements in high-throughput sequencing technologies and bioinformatic analyses have enabled high-resolution analysis of local and peripheral adaptive immune repertoires in the setting of central nervous system malignancies. These robust methods offer exciting new techniques for quantifying antitumor immune responses that can be harnessed as biomarkers for response to immune treatment or as a means to design cell therapy strategies. In the future, immunogenomic studies will be critical to understanding the genomic basis of immunotherapy response and resistance and to inform the next generation of immunotherapy trials.
- Published
- 2022
92. Contributors
- Author
-
April Bell, Orin Bloch, Lakshmi Bollu, E. Antonio Chiocca, Pavlina Chuntova, Andrew T. Coxon, Gavin P. Dunn, Susannah G. Ellsworth, Peter E. Fecci, Ryan Gilbert, Stuart A. Grossman, Aden P. Haskell-Mendoza, Lan B. Hoang-Minh, Michael Jin, Miri Kim, Erik Ladomersky, Kristen L. Lauing, Gordon Li, Connor J. Liu, Selena Lorrey, Duane A. Mitchell, Hideho Okada, Jessica Waibl Polania, Erik Rabin, Isaac H. Solomon, Derek A. Wainwright, Payal B. Watchmaker, Daniel Wilkinson, Adela Wu, and Lijie Zhai
- Published
- 2022
93. Multivariate analysis of associations between clinical sequencing and outcome in glioblastoma
- Author
-
Peter H Yang, Yu Tao, Jingqin Luo, Mounica Paturu, Hsiang-Chih Lu, Shakti Ramkissoon, Jonathan W Heusel, Eric C Leuthardt, Michael R Chicoine, Joshua L Dowling, Gavin P Dunn, Eric Duncavage, Sonika Dahiya, Arindam R Chattherjee, and Albert H Kim
- Abstract
Background Many factors impact survival in patients with glioblastoma, including age, Karnofsky Performance Status, postoperative chemoradiation, IDH1/2 mutation status, MGMT promoter methylation status, and extent of resection. High-throughput next-generation sequencing is a widely available diagnostic tool, but the independent impact of tumors harboring specific mutant genes on survival and the efficacy of extent of resection are not clear. Methods We utilized a widely available diagnostic platform (FoundationOne CDx) to perform high-throughput next-generation sequencing on 185 patients with newly diagnosed glioblastoma in our tertiary care center. We performed multivariate analysis to control for clinical parameters with known impact on survival to elucidate the independent prognostic value of prevalent mutant genes and the independent impact of gross total resection. Results When controlling for factors with known prognostic significance including IDH1/2 mutation and after multiple comparisons analysis, CDKN2B and EGFR mutations were associated with reduced overall survival while PTEN mutation was associated with improved overall survival. Gross total resection, compared to other extent of resection, was associated with improved overall survival in patients with tumors harboring mutations in CDKN2A, CDKN2B, EGFR, PTEN, TERT promoter, and TP53. All patients possessed at least one of these 6 mutant genes. Conclusions This study verifies the independent prognostic value of several mutant genes in glioblastoma. Six commonly found mutant genes were associated with improved survival when gross total resection was achieved. Thus, even when accounting for known predictors of survival and multiple mutant gene comparisons, extent of resection continues to be strongly associated with survival.
- Published
- 2022
94. TCR-engineered adoptive cell therapy effectively treats intracranial murine glioblastoma
- Author
-
Maximilian O Schaettler, Rupen Desai, Anthony Z Wang, Alexandra J Livingstone, Dale K Kobayashi, Andrew T Coxon, Jay A Bowman-Kirigin, Connor J Liu, Mao Li, Diane E Bender, Michael J White, David M Kranz, Tanner M Johanns, and Gavin P Dunn
- Subjects
Pharmacology ,Cancer Research ,Oncology ,Immunology ,Molecular Medicine ,Immunology and Allergy - Abstract
BackgroundAdoptive cellular therapies with chimeric antigen receptor T cells have revolutionized the treatment of some malignancies but have shown limited efficacy in solid tumors such as glioblastoma and face a scarcity of safe therapeutic targets. As an alternative, T cell receptor (TCR)–engineered cellular therapy against tumor-specific neoantigens has generated significant excitement, but there exist no preclinical systems to rigorously model this approach in glioblastoma.MethodsWe employed single-cell PCR to isolate a TCR specific for the Imp3D81Nneoantigen (mImp3) previously identified within the murine glioblastoma model GL261. This TCR was used to generate the Mutant Imp3-Specific TCR TransgenIC (MISTIC) mouse in which all CD8 T cells are specific for mImp3. The therapeutic efficacy of neoantigen-specific T cells was assessed through a model of cellular therapy consisting of the transfer of activated MISTIC T cells and interleukin 2 into lymphodepleted tumor-bearing mice. We employed flow cytometry, single-cell RNA sequencing, and whole-exome and RNA sequencing to examine the factors underlying treatment response.ResultsWe isolated and characterized the 3×1.1C TCR that displayed a high affinity for mImp3 but no wild-type cross-reactivity. To provide a source of mImp3-specific T cells, we generated the MISTIC mouse. In a model of adoptive cellular therapy, the infusion of activated MISTIC T cells resulted in rapid intratumoral infiltration and profound antitumor effects with long-term cures in a majority of GL261-bearing mice. The subset of mice that did not respond to the adoptive cell therapy showed evidence of retained neoantigen expression but intratumoral MISTIC T cell dysfunction. The efficacy of MISTIC T cell therapy was lost in mice bearing a tumor with heterogeneous mImp3 expression, showcasing the barriers to targeted therapy in polyclonal human tumors.ConclusionsWe generated and characterized the first TCR transgenic against an endogenous neoantigen within a preclinical glioma model and demonstrated the therapeutic potential of adoptively transferred neoantigen-specific T cells. The MISTIC mouse provides a powerful novel platform for basic and translational studies of antitumor T-cell responses in glioblastoma.
- Published
- 2023
95. Impact of CD4 T cells on intratumoral CD8 T-cell exhaustion and responsiveness to PD-1 blockade therapy in mouse brain tumors
- Author
-
Saad M Khan, Rupen Desai, Andrew Coxon, Alexandra Livingstone, Gavin P Dunn, Allegra Petti, and Tanner M Johanns
- Subjects
CD4-Positive T-Lymphocytes ,Pharmacology ,Cancer Research ,Brain Neoplasms ,Programmed Cell Death 1 Receptor ,Immunology ,CD8-Positive T-Lymphocytes ,T-Cell Exhaustion ,Mice ,Oncology ,Tumor Microenvironment ,Animals ,Molecular Medicine ,Immunology and Allergy ,Female ,Glioblastoma - Abstract
BackgroundGlioblastoma is a fatal disease despite aggressive multimodal therapy. PD-1 blockade, a therapy that reinvigorates hypofunctional exhausted CD8 T cells (Tex) in many malignancies, has not shown efficacy in glioblastoma. Loss of CD4 T cells can lead to an exhausted CD8 T-cell phenotype, and terminally exhausted CD8 T cells (Texterm) do not respond to PD-1 blockade. GL261 and CT2A are complementary orthotopic models of glioblastoma. GL261 has a functional CD4 T-cell compartment and is responsive to PD-1 blockade; notably, CD4 depletion abrogates this survival benefit. CT2A is composed of dysfunctional CD4 T cells and is PD-1 blockade unresponsive. We leverage these models to understand the impact of CD4 T cells on CD8 T-cell exhaustion and PD-1 blockade sensitivity in glioblastoma.MethodsSingle-cell RNA sequencing was performed on flow sorted tumor-infiltrating lymphocytes from female C57/BL6 mice implanted with each model, with and without PD-1 blockade therapy. CD8+and CD4+T cells were identified and separately analyzed. Survival analyses were performed comparing PD-1 blockade therapy, CD40 agonist or combinatorial therapy.ResultsThe CD8 T-cell compartment of the models is composed of heterogenous CD8 Texsubsets, including progenitor exhausted CD8 T cells (Texprog), intermediate Tex, proliferating Tex, and Texterm. GL261 is enriched with the PD-1 responsive Texprogsubset relative to the CT2A and CD4-depleted GL261 models, which are composed predominantly of the PD-1 blockade refractory Textermsubset. Analysis of the CD4 T-cell compartments revealed that the CT2A microenvironment is enriched with a suppressive Tregsubset and an effector CD4 T-cell subset that expresses an inhibitory interferon-stimulated (Isc) signature. Finally, we demonstrate that addition of CD40 agonist to PD-1 blockade therapy improves survival in CT2A tumor-bearing mice.ConclusionsHere, we describe that dysfunctional CD4 T cells are associated with terminal CD8 T-cell exhaustion, suggesting CD4 T cells impact PD-1 blockade efficacy by controlling the severity of exhaustion. Given that CD4 lymphopenia is frequently observed in patients with glioblastoma, this may represent a basis for resistance to PD-1 blockade. We demonstrate that CD40 agonism may circumvent a dysfunctional CD4 compartment to improve PD-1 blockade responsiveness, supporting a novel synergistic immunotherapeutic approach.
- Published
- 2022
96. Circulating Immune Cell and Outcome Analysis from the Phase II Study of PD-L1 Blockade with Durvalumab for Newly Diagnosed and Recurrent Glioblastoma
- Author
-
Lakshmi Nayak, Nathan Standifer, Jorg Dietrich, Jennifer L. Clarke, Gavin P. Dunn, Michael Lim, Timothy Cloughesy, Hui K. Gan, Elizabeth Flagg, Elizabeth George, Sarah Gaffey, Julia Hayden, Christina Holcroft, Patrick Y. Wen, Mary Macri, Andrew J. Park, Toni Ricciardi, Aileen Ryan, Paul Schwarzenberger, Ralph Venhaus, Melissa de los Reyes, Nicholas M. Durham, Todd Creasy, Raymond Y. Huang, Thomas Kaley, and David A. Reardon
- Subjects
Cancer Research ,Tumor ,Oncology and Carcinogenesis ,Antibodies, Monoclonal ,Antibodies ,B7-H1 Antigen ,Dexamethasone ,Article ,Brain Disorders ,Brain Cancer ,Bevacizumab ,Neoplasm Recurrence ,Rare Diseases ,Ki-67 Antigen ,Oncology ,Local ,Clinical Research ,Monoclonal ,Biomarkers, Tumor ,Humans ,Oncology & Carcinogenesis ,Neoplasm Recurrence, Local ,Glioblastoma ,Biomarkers ,Cancer - Abstract
Purpose: PD-L1 is upregulated in glioblastoma and supports immunosuppression. We evaluated PD-L1 blockade with durvalumab among glioblastoma cohorts and investigated potential biomarkers. Patients and Methods: MGMT unmethylated newly diagnosed patients received radiotherapy plus durvalumab (cohort A; n = 40). Bevacizumab-naïve, recurrent patients received durvalumab alone (cohort B; n = 31) or in combination with standard bevacizumab (cohort B2; n = 33) or low-dose bevacizumab (cohort B3; n = 33). Bevacizumab-refractory patients received durvalumab plus bevacizumab (cohort C; n = 22). Primary endpoints were: OS-12 (A), PFS-6 (B, B2, B3), and OS-6 (C). Exploratory biomarkers included: a systematic, quantitative, and phenotypic evaluation of circulating immune cells; tumor mutational burden (TMB); and tumor immune activation signature (IAS). Results: No cohort achieved the primary efficacy endpoint. Outcome was comparable among recurrent, bevacizumab-naïve cohorts. No unexpected toxicities were observed. A widespread reduction of effector immune cell subsets was noted among recurrent patients compared with newly diagnosed patients that was partially due to dexamethasone use. A trend of increased CD8+Ki67+ T cells at day 15 was noted among patients who achieved the primary endpoint and were not on dexamethasone. Neither TMB nor IAS predicted outcome. Conclusions: Patients with recurrent glioblastoma have markedly lower baseline levels of multiple circulating immune cell subsets compared with newly diagnosed patients. An early increase in systemic Ki67+CD8+ cells may warrant further evaluation as a potential biomarker of therapeutic benefit among patients with glioblastoma undergoing checkpoint therapy. Dexamethasone decreased immune cell subsets. PD-L1 blockade and combination with standard or reduced dose bevacizumab was ineffective.
- Published
- 2021
97. CTIM-26. PHASE I/II STUDY OF THE COMBINATION OF PEMBROLIZUMAB (MK-3475) AND LASER INTERSTITIAL THERMAL THERAPY (LITT) IN RECURRENT GLIOBLASTOMA
- Author
-
Eric C. Leuthardt, Ashley Ghinaseddin, Albert H. Kim, Tanner M. Johanns, David Tran, Alice Zhou, Omar Butt, Jiayi Huang, Gavin P. Dunn, Jingxia Liu, Maryam Rahman, George Ansstas, Grayson Talcott, Jian Campian, Ruth Katumba, William Leidig, and Milan G. Chheda
- Subjects
Cancer Research ,Materials science ,Bevacizumab ,business.industry ,Recurrent glioblastoma ,Pembrolizumab ,26th Annual Meeting & Education Day of the Society for Neuro-Oncology ,medicine.disease ,Phase i ii ,Oncology ,Laser Interstitial Thermal Therapy ,Glioma ,medicine ,Neurology (clinical) ,Nuclear medicine ,business ,Anaplastic astrocytoma ,medicine.drug ,Glioblastoma - Abstract
BACKGROUND The blood brain barrier (BBB) remains a potential barrier to central nervous system (CNS) penetration of novel immunotherapies in recurrent glioblastoma (rGBM). Laser interstitial thermal therapy (LITT) was recently demonstrated to induce BBB disruption. When combined with anti-PD1 blockade, LITT may therefore potentiate host T-cell mediated cytotoxicity. This phase I/II study aims to evaluate the safety and efficacy of combining LITT and the PD-1 inhibitor pembrolizumab for rGBM. METHODS Phase I treated eligible bevacizumab-naïve high grade glioma patients with LITT and the anti-PD1 inhibitor pembrolizumab at 3 dose levels (100, 150, and 200 mg IV Q3W; 3 patients at each level), while phase II was restricted to rGBM patients only with the recommended phase II dose (RP2D) of pembrolizumab. Phase II was initially designed to randomize rGBM to either pembrolizumab or pembrolizumab+LITT but was later amended to receive only pembrolizumab+LITT after 16 patients were randomized (10 pembrolizumab+LITT arm, 6 pembrolizumab-alone arm). Progression-free survival (PFS) and overall survival (OS) were evaluated using the Kaplan Meier method, and adverse events (AE) documented. RESULTS Phase I enrolled 9 patients (7 rGBM and 2 anaplastic astrocytomas, 33% IDH-mut, 44% MGMTp-methylated) with no dose limiting toxicities (DLT), prompting selection of 200mg Q3W as the RP2D. Phase II interim analysis included 34 rGBM patients (9% IDH-mut, 50% MGMTp-methylated; 6 receiving pembrolizumab alone and 28 pembrolizumab+LITT) plus two Phase I rGBM patients who received RP2D. On per-protocol analysis, pembrolizumab+LITT cohort had improved PFS (median PFS 10.5 months vs 2.1 months) and OS (median OS 11.4 months vs 5.2 months) than pembrolizumab alone. A single treatment-related grade 3 AE was noted (respiratory infection). CONCLUSION: LITT may be safely combined with pembrolizumab for rGBM with promising clinical outcomes on interim analysis. Enrollment for Phase II, correlative studies, and continued AE documentation are ongoing. Clinical Trial ID NCT02311582.
- Published
- 2021
98. Competitive binding of E3 ligases TRIM26 and WWP2 controls SOX2 in glioblastoma
- Author
-
Gavin P. Dunn, Albert H. Kim, Amit D. Gujar, Nima Mosammaparast, Allegra A. Petti, Hiroko Yano, Mounica Paturu, Wei Yang, Daniel Hafez, Tatenda Mahlokozera, Rukayat Taiwo, Xuan Qu, Afshin Salehi, Diane D. Mao, Hao Chen, Bhuvic Patel, and Patrick A. DeSouza
- Subjects
Proteomics ,endocrine system ,Ubiquitylation ,Science ,Ubiquitin-Protein Ligases ,General Physics and Astronomy ,WWP2 ,Mice, SCID ,B30.2-SPRY Domain ,Binding, Competitive ,Article ,General Biochemistry, Genetics and Molecular Biology ,Tripartite Motif Proteins ,Mice ,stomatognathic system ,Ubiquitin ,SOX2 ,Mice, Inbred NOD ,Cancer stem cell ,Animals ,Humans ,Transcription factor ,Gene knockdown ,Multidisciplinary ,biology ,Brain Neoplasms ,Cancer stem cells ,SOXB1 Transcription Factors ,fungi ,Ubiquitination ,General Chemistry ,Cell biology ,Ubiquitin ligase ,HEK293 Cells ,Gene Knockdown Techniques ,embryonic structures ,biology.protein ,Female ,sense organs ,biological phenomena, cell phenomena, and immunity ,Stem cell ,Glioblastoma - Abstract
The pluripotency transcription factor SOX2 is essential for the maintenance of glioblastoma stem cells (GSC), which are thought to underlie tumor growth, treatment resistance, and recurrence. To understand how SOX2 is regulated in GSCs, we utilized a proteomic approach and identified the E3 ubiquitin ligase TRIM26 as a direct SOX2-interacting protein. Unexpectedly, we found TRIM26 depletion decreased SOX2 protein levels and increased SOX2 polyubiquitination in patient-derived GSCs, suggesting TRIM26 promotes SOX2 protein stability. Accordingly, TRIM26 knockdown disrupted the SOX2 gene network and inhibited both self-renewal capacity as well as in vivo tumorigenicity in multiple GSC lines. Mechanistically, we found TRIM26, via its C-terminal PRYSPRY domain, but independent of its RING domain, stabilizes SOX2 protein by directly inhibiting the interaction of SOX2 with WWP2, which we identify as a bona fide SOX2 E3 ligase in GSCs. Our work identifies E3 ligase competition as a critical mechanism of SOX2 regulation, with functional consequences for GSC identity and maintenance., SOX2 is required for the maintenance of glioblastoma stem cells (GSCs). Here the authors identify that the RING family E3 ubiquitin ligase TRIM26 promotes SOX2 stability in a non-canonical ligase-independent manner and thus, increases the tumorigenicity of GSCs.
- Published
- 2021
99. 777 Personalized DNA vaccine in combination with plasmid encoded IL-12 for the treatment of a patient with anaplastic astrocytoma
- Author
-
Niranjan Y. Sardesai, Alfredo Perales-Puchalt, Tanner M. Johanns, Sarah Rochestie, Neil Cooch, Joann Peters, and Gavin P. Dunn
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,T cell ,Immunology ,Epitope ,DNA vaccination ,Antigen ,Internal medicine ,medicine ,Immunology and Allergy ,RC254-282 ,Pharmacology ,Temozolomide ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Vaccination ,medicine.anatomical_structure ,Molecular Medicine ,business ,Adjuvant ,Anaplastic astrocytoma ,medicine.drug - Abstract
BackgroundTumor neoantigens are epitopes derived from tumor-specific mutations. Such mutations can be incorporated in personalized vaccines to prime T cell responses against tumor specific antigens. DNA vaccines delivered with electroporation have recently shown strong CD8 and CD4 T cell responses in clinical trials. In preclinical studies, DNA-encoded neoantigen vaccines have shown induction of CD8 T cells against 50% of predicted high affinity epitopes with the ability to impact tumor growth.MethodsTwo resection samples from a patient with IDH+ MGMT-methylated anaplastic astrocytoma were subject to whole exome and transcriptome sequencing. Epitopes derived from 27 neoantigens and 3 shared tumor-associated antigens were prioritized and included in a personalized vaccine. The patient was treated with surgery, radiotherapy and temozolomide starting June 2018 and received the first dose of the personalized vaccine in June 2019 under a compassionate use single patient IND application with the FDA.ResultsAs of July 23rd, 2021, the patient has received 11 doses of the DNA personalized vaccine. No serious adverse events have been reported. Related adverse events are limited to grade 1 injection site reactions. The patient remains progression-free 37 months after surgery and 25 months after starting vaccination. Three weeks following the 3rd dose, a hyperintense image on the tumor bed was identified, which disappeared on the following MRI, 2 weeks following dose 5, being catalogued as pseudo progression. Ex vivo ELISpot have identified T cell responses to 28/30 epitopes (93.3%), including 25/27 (92.6%) neoantigens and 3/3 (100%) shared antigens. Flow cytometry analysis has determined that T cell responses are 92.3% CD8 and 69.2% CD4 (30.8% CD8 only; 61.5% both CD8 and CD4; and 7.7% CD4 only).ConclusionsThis compassionate use treatment in an adjuvant setting demonstrates manufacturing feasibility, safety, tolerability, immunogenicity, and suggests potential for persistent clinical response of DNA encoded personalized vaccines. The data supports further investigation of DNA-encoded personalized vaccines into newly diagnosed high-grade gliomas.Ethics ApprovalThe study was approved by Washington University's IRB. The participant gave informed consent before taking part in the study.
- Published
- 2021
100. Considerations for personalized neoantigen vaccination in Malignant glioma
- Author
-
Gavin P. Dunn, Ngima Sherpa, Jimmy Manyanga, and Tanner M. Johanns
- Subjects
Antigens, Neoplasm ,Neoplasms ,Vaccination ,Pharmaceutical Science ,Humans ,Immunologic Factors ,Glioma ,Immunotherapy ,Glioblastoma ,Cancer Vaccines - Abstract
Malignant gliomas are the most common primary brain cancer diagnosed and still carry a poor prognosis despite aggressive multimodal management. Despite the continued advances in immunotherapy for other cancer types, however, there remain no FDA approved immunotherapies for cancers such as glioblastoma. OF the many approaches being explored, cancer vaccine programs are undergoing a renaissance due to the technological advances and personalized nature of their contemporary design. Neoantigen vaccines are a form of immunotherapy involving the use of DNA, mRNA, and proteins derived from non-synonymous mutations identified in patient tumor tissue samples to stimulate tumor-specific T-cell reactivity leading to enhance tumor targeting. In the last several years, the study of neoantigens as a therapeutic target has increased, with the routine workflow implementation of comprehensive next generation sequencing and in silico peptide binding prediction algorithms. Several neoantigen vaccine platforms are being evaluated in clinical trials for malignancies including melanoma, pancreatic cancer, breast cancer, lung cancer, and glioblastoma, among others. In this review, we will review the concept of neoantigen discovery using cancer immunogenomics approaches in glioblastoma and explore the disease-specific issues being addressed in the design of effective personalized cancer vaccine strategies.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.