100 results on '"Venu G Pillarisetty"'
Search Results
2. IL-15 mediated expansion of rare durable memory T cells following adoptive cellular therapy
- Author
-
Robin L Jones, Erik A Farrar, Jianhong Cao, Venu G Pillarisetty, Stanley R Riddell, Jean Campbell, Brett A Schroeder, Ralph Graeme Black, Shihong Zhang, Karan Kohli, Robert H Pierce, Lu Yao, Theodore Scott Nowicki, Heather Sloan, Dawn Stief, Lee D Cranmer, Douglas S Hawkins, and Edward Y Kim
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background Synovial sarcoma (SS) and myxoid/round cell liposarcoma (MRCL) are ideal solid tumors for the development of adoptive cellular therapy (ACT) targeting NY-ESO-1, as a high frequency of tumors homogeneously express this cancer-testes antigen. Data from early phase clinical trials have shown antitumor activity after the adoptive transfer of NY-ESO-1–specific T cells. In these studies, persistence of NY-ESO-1 specific T cells is highly correlated with response to ACT, but patients often continue to have detectable transferred cells in their peripheral blood following progression.Method We performed a phase I clinical trial evaluating the safety of NY-ESO-1–specific endogenous T cells (ETC) following cyclophosphamide conditioning. Peripheral blood mononuclear cells (PBMCs) from treated patients were evaluated by flow cytometry and gene expression analysis as well as through ex vivo culture assays with and without IL-15.Results Four patients were treated in a cohort using ETC targeting NY-ESO-1 following cyclophosphamide conditioning. Treatment was well tolerated without significant toxicity, but all patients ultimately had disease progression. In two of four patients, we obtained post-treatment tumor tissue and in both, NY-ESO-1 antigen was retained despite clear detectable persisting NY-ESO-1–specific T cells in the peripheral blood. Despite a memory phenotype, these persisting cells lacked markers of proliferation or activation. However, in ex vivo culture assays, they could be induced to proliferate and kill tumor using IL-15. These results were also seen in PBMCs from two patients who received gene-engineered T-cell receptor–based products at other centers.Conclusions ETC targeting NY-ESO-1 with single-agent cyclophosphamide alone conditioning was well tolerated in patients with SS and those with MRCL. IL-15 can induce proliferation and activity in persisting NY-ESO-1–specific T cells even in patients with disease progression following ACT. These results support future work evaluating whether IL-15 could be incorporated into ACT trials post-infusion or at the time of progression.
- Published
- 2021
- Full Text
- View/download PDF
3. Pancreatic paraganglioma mimicking pancreatic neuroendocrine tumor
- Author
-
Arezou Abbasi, Kristina M Wakeman, and Venu G Pillarisetty
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Extra-adrenal paragangliomas are rare tumors arising from the chromaffin cells of the autonomic nervous system. Retroperitoneal paragangliomas may present as a pancreatic mass. We present a case of a 61-year-old woman with an incidentally found pancreatic mass (7.2 × 6.5 cm) in the CT scan. EUS- guided FNA result was compatible with pancreatic neuroendocrine tumor. Patient underwent pancreaticoduodenectomy and histopathologic assessment revealed the mass was an extra-adrenal paraganglioma. Preoperative diagnosis of pancreatic paragangliomas can be challenging due to imaging and histopathologic similarities with pancreatic neuroendocrine tumors.
- Published
- 2020
- Full Text
- View/download PDF
4. Genetically engineered macrophages persist in solid tumors and locally deliver therapeutic proteins to activate immune responses
- Author
-
Michael C Jensen, Venu G Pillarisetty, Katherine J Brempelis, Courtney M Cowan, Shannon A Kreuser, Kevin P Labadie, Brooke M Prieskorn, Nicole A P Lieberman, Chibawanye I Ene, Kara W Moyes, Harrison Chinn, Kole R DeGolier, Lisa R Matsumoto, Sara K Daniel, Jason K Yokoyama, Amira D Davis, Virginia J Hoglund, Kimberly S Smythe, Stephanie D Balcaitis, Richard G Ellenbogen, Jean S Campbell, Robert H Pierce, Eric C Holland, and Courtney A Crane
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background Though currently approved immunotherapies, including chimeric antigen receptor T cells and checkpoint blockade antibodies, have been successfully used to treat hematological and some solid tumor cancers, many solid tumors remain resistant to these modes of treatment. In solid tumors, the development of effective antitumor immune responses is hampered by restricted immune cell infiltration and an immunosuppressive tumor microenvironment (TME). An immunotherapy that infiltrates and persists in the solid TME, while providing local, stable levels of therapeutic to activate or reinvigorate antitumor immunity could overcome these challenges faced by current immunotherapies.Methods Using lentivirus-driven engineering, we programmed human and murine macrophages to express therapeutic payloads, including Interleukin (IL)-12. In vitro coculture studies were used to evaluate the effect of genetically engineered macrophages (GEMs) secreting IL-12 on T cells and on the GEMs themselves. The effects of IL-12 GEMs on gene expression profiles within the TME and tumor burden were evaluated in syngeneic mouse models of glioblastoma and melanoma and in human tumor slices isolated from patients with advanced gastrointestinal malignancies.Results Here, we present a cellular immunotherapy platform using lentivirus-driven genetic engineering of human and mouse macrophages to constitutively express proteins, including secreted cytokines and full-length checkpoint antibodies, as well as cytoplasmic and surface proteins that overcomes these barriers. GEMs traffic to, persist in, and express lentiviral payloads in xenograft mouse models of glioblastoma, and express a non-signaling truncated CD19 surface protein for elimination. IL-12-secreting GEMs activated T cells and induced interferon-gamma (IFNγ) in vitro and slowed tumor growth resulting in extended survival in vivo. In a syngeneic glioblastoma model, IFNγ signaling cascades were also observed in mice treated with mouse bone-marrow-derived GEMs secreting murine IL-12. These findings were reproduced in ex vivo tumor slices comprised of intact MEs. In this setting, IL-12 GEMs induced tumor cell death, chemokines and IFNγ-stimulated genes and proteins.Conclusions Our data demonstrate that GEMs can precisely deliver titratable doses of therapeutic proteins to the TME to improve safety, tissue penetrance, targeted delivery and pharmacokinetics.
- Published
- 2020
- Full Text
- View/download PDF
5. Standardization of perioperative care facilitates safe discharge by postoperative day five after pancreaticoduodenectomy.
- Author
-
Sara K Daniel, Lucas W Thornblade, Gary N Mann, James O Park, and Venu G Pillarisetty
- Subjects
Medicine ,Science - Abstract
IntroductionPancreaticoduodenectomy is a complex surgical procedure associated with high morbidity and prolonged length of stay. Enhanced recovery after surgery principles have reduced complications rate and length of stay for multiple types of operations. We hypothesized that implementation of a standardized perioperative care pathway would facilitate safe discharge by five days after pancreaticoduodenectomy.MethodsWe performed a retrospective cohort study of patients undergoing pancreaticoduodenectomy 18 months prior to and 18 months following implementation of a perioperative care pathway at a quaternary center performing high volume pancreatic surgery.ResultsA total of 145 patients underwent pancreaticoduodenectomy (mean age 63 ± 10 years, 52% female), 81 before and 64 following pathway implementation, and the groups were similar in terms of preoperative comorbidities. The percentage of patients discharged within 5 days of surgery increased from 36% to 64% following pathway implementation (p = 0.001), with no observed differences in post-operative serious adverse events (p = 0.34), pancreatic fistula grade B or C (p = 0.28 and p = 0.27 respectively), or delayed gastric emptying (p = 0.46). Multivariate regression analysis showed length of stay ≤5 days three times more likely after pathway implementation. Rates of readmission within 30 days (20% pre- vs. 22% post-pathway (p = 0.75)) and 90 days (27% pre- vs. 36% post-pathway (p = 0.27)) were unchanged after pathway implementation, and were no different between patients discharged before or after day 5 at both 30 days (19% ≤5 days vs. 23% ≥ 6 days (p = 0.68)) and 90 days (32% ≤5 days vs. 30% ≥ 6 days (p = 0.81)).ConclusionsStandardizing perioperative care via enhanced recovery protocols for patients undergoing pancreaticoduodenectomy facilitates safe discharge by post-operative day five.
- Published
- 2018
- Full Text
- View/download PDF
6. FOXP3+ lymphocyte density in pancreatic cancer correlates with lymph node metastasis.
- Author
-
Yongjian Jiang, Zunguo Du, Feng Yang, Yang Di, Ji Li, Zhongwen Zhou, Venu G Pillarisetty, and Deliang Fu
- Subjects
Medicine ,Science - Abstract
To determine if the density of FOXP3+ lymphocytes in primary tumors and lymph nodes in pancreatic cancer correlates with the presence of lymph node metastases.FOXP3+ lymphocyte density in primary pancreatic cancer tissue and draining lymph nodes was measured using immunohistochemistry. We analyzed the clinical and pathological aspects associated with the accumulation of FOXP3+ lymphocytes in pancreatic cancer. We also analyzed the correlation of density of FOXP3+ lymphocytes in lymph nodes with the nodal status and distance from the primary tumor.FOXP3+ lymphocyte density in pancreatic cancer was significantly higher than in paratumoral pancreatic tissue. The density of FOXP3+ lymphocytes in local tumor tissue correlated significantly with the histological grade and overall lymph node status. Furthermore, FOXP3+ lymphocyte density was significantly higher in positive lymph nodes than in negative ones, while it had no correlation with the distance of the lymph node from the primary tumor.FOXP3+ lymphocyte density in primary tumor tissue in patients with pancreatic cancer correlates with lymph node metastasis. Lymph nodes containing metastases having higher FOXP3+ lymphocyte densities than do negative lymph nodes.
- Published
- 2014
- Full Text
- View/download PDF
7. Pancreatic ductal adenocarcinoma contains an effector and regulatory immune cell infiltrate that is altered by multimodal neoadjuvant treatment.
- Author
-
Kendall C Shibuya, Vikas K Goel, Wei Xiong, Jonathan G Sham, Seth M Pollack, Allison M Leahy, Samuel H Whiting, Matthew M Yeh, Cassian Yee, Stanley R Riddell, and Venu G Pillarisetty
- Subjects
Medicine ,Science - Abstract
OBJECTIVE:The immune response to pancreatic ductal adenocarcinoma (PDA) may play a role in defining its uniquely aggressive biology; therefore, we sought to clearly define the adaptive immune infiltrate in PDA. DESIGN:We used immunohistochemistry and flow cytometry to characterize the immune infiltrate in human PDA and compared our findings to the patients' peripheral blood. RESULTS:In contrast to the myeloid cell predominant infiltrate seen in murine models, T cells comprised the majority of the hematopoietic cell component of the tumor stroma in human PDA. Most intratumoral CD8+ T cells exhibited an antigen-experienced effector memory cell phenotype and were capable of producing IFN-γ. CD4+ regulatory T cells (Treg) and IL-17 producing T helper cells were significantly more prevalent in tumor than in blood. Consistent with the association with reduced survival in previous studies, we observed higher frequencies of both myeloid cells and Treg in poorly differentiated tumors. The majority of intratumoral T cells expressed the co-inhibitory receptor programmed death-1 (PD-1), suggesting one potential mechanism through which PDA may evade antitumor immunity. Successful multimodal neoadjuvant therapy altered the immunoregulatory balance and was associated with reduced infiltration of both myeloid cells and Treg. CONCLUSION:Our data show that human PDA contains a complex mixture of inflammatory and regulatory immune cells, and that neoadjuvant therapy attenuates the infiltration of intratumoral cells associated with immunosuppression and worsened survival.
- Published
- 2014
- Full Text
- View/download PDF
8. A phase II trial of lanreotide for the prevention of postoperative pancreatic fistula
- Author
-
Venu G. Pillarisetty, Arezou Abbasi, James O. Park, and Jonathan G. Sham
- Subjects
Pancreatic Fistula ,Pancreatectomy ,Postoperative Complications ,Hepatology ,Risk Factors ,Gastroenterology ,Humans ,Somatostatin ,Pancreaticoduodenectomy ,Retrospective Studies - Abstract
Clinically relevant postoperative pancreatic fistula (CR-POPF) is a significant contributor to morbidity after pancreatectomy. Somatostatin analogues have shown variable efficacy in the prevention of CR-POPF. Lanreotide is a somatostatin analogue ideally suited for perioperative use due to its long half-life and favorable side effect profile.We conducted a phase II single-arm trial of a single dose of preoperative lanreotide (120 mg) in patients undergoing either pancreaticoduodenectomy (PD) or distal pancreatectomy (DP). The primary outcome was development of CR-POPF or intra-abdominal abscess. Secondary outcomes included biochemical leak and overall morbidity.A total of 98 patients completed the study. Sixty-two underwent PD (63.3%) and 36 underwent DP (36.7%). The primary outcome was observed in eight (8%) patients in the overall cohort, one from the DP group and seven from the PD group. Biochemical leak was detected in 12 (12.2%) patients in the overall cohort. Twenty-seven (27.5%) patients developed complications, of which 14 (14.2%) were major complications. Drug-related adverse events were limited to mild skin reactions in two (2%) patients.Patients who received preoperative lanreotide developed CR-POPF at rates significantly lower than historical controls or published literature. This provides strong justification for a randomized controlled trial.
- Published
- 2022
9. Disrupting glutamine metabolic pathways to sensitize gemcitabine-resistant pancreatic cancer
- Author
-
Ru Chen, Lisa A Lai, Yumi Sullivan, Melissa Wong, Lei Wang, Jonah Riddell, Linda Jung, Venu G. Pillarisetty, Teresa A. Brentnall, and Sheng Pan
- Subjects
Medicine ,Science - Abstract
Abstract Pancreatic cancer is a lethal disease with poor prognosis. Gemcitabine has been the first line systemic treatment for pancreatic cancer. However, the rapid development of drug resistance has been a major hurdle in gemcitabine therapy leading to unsatisfactory patient outcomes. With the recent renewed understanding of glutamine metabolism involvement in drug resistance and immuno-response, we investigated the anti-tumor effect of a glutamine analog (6-diazo-5-oxo-L-norleucine) as an adjuvant treatment to sensitize chemoresistant pancreatic cancer cells. We demonstrate that disruption of glutamine metabolic pathways improves the efficacy of gemcitabine treatment. Such a disruption induces a cascade of events which impacts glycan biosynthesis through Hexosamine Biosynthesis Pathway (HBP), as well as cellular redox homeostasis, resulting in global changes in protein glycosylation, expression and functional effects. The proteome alterations induced in the resistant cancer cells and the secreted exosomes are intricately associated with the reduction in cell proliferation and the enhancement of cancer cell chemosensitivity. Proteins associated with EGFR signaling, including downstream AKT-mTOR pathways, MAPK pathway, as well as redox enzymes were downregulated in response to disruption of glutamine metabolic pathways.
- Published
- 2017
- Full Text
- View/download PDF
10. Supplementary Data from Systemic Interferon-γ Increases MHC Class I Expression and T-cell Infiltration in Cold Tumors: Results of a Phase 0 Clinical Trial
- Author
-
Seth M. Pollack, Robin L. Jones, Stanley R. Riddell, Robert H. Pierce, Cassian Yee, Brian A. Van Tine, Lee D. Cranmer, Venu G. Pillarisetty, Qianchuan He, Sydney M. Spadinger, Lu Yao, R. Graeme Black, Karan Kohli, and Shihong Zhang
- Abstract
Supplementary Tables 1-3 and Supplementary Tables 1-6
- Published
- 2023
11. Data from Systemic Interferon-γ Increases MHC Class I Expression and T-cell Infiltration in Cold Tumors: Results of a Phase 0 Clinical Trial
- Author
-
Seth M. Pollack, Robin L. Jones, Stanley R. Riddell, Robert H. Pierce, Cassian Yee, Brian A. Van Tine, Lee D. Cranmer, Venu G. Pillarisetty, Qianchuan He, Sydney M. Spadinger, Lu Yao, R. Graeme Black, Karan Kohli, and Shihong Zhang
- Abstract
Interferon-γ (IFNγ) has been studied as a cancer treatment with limited evidence of clinical benefit. However, it could play a role in cancer immunotherapy combination treatments. Despite high expression of immunogenic cancer–testis antigens, synovial sarcoma (SS) and myxoid/round cell liposarcoma (MRCL) have a cold tumor microenvironment (TME), with few infiltrating T cells and low expression of major histocompatibility complex class I (MHC-I). We hypothesized that IFNγ treatment could drive inflammation in a cold TME, facilitating further immunotherapy. We conducted a phase 0 clinical trial treating 8 SS or MRCL patients with weekly systemic IFNγ. We performed pre- and posttreatment biopsies. IFNγ changed the SS and MRCL TME, inducing tumor-surface MHC-I expression and significant T-cell infiltration (P < 0.05). Gene-expression analysis suggested increased tumor antigen presentation and less exhausted phenotypes of the tumor-infiltrating T cells. Newly emergent antigen-specific humoral and/or T-cell responses were found in 3 of 7 evaluable patients. However, increased expression of PD-L1 was observed on tumor-infiltrating myeloid cells and in some cases tumor cells. These findings suggest that systemic IFNγ used to convert SS and MRCL into “hot” tumors will work in concert with anti–PD-1 therapy to provide patient benefit.
- Published
- 2023
12. Supplementary Figure 6 from Mobilization of CD8+ T Cells via CXCR4 Blockade Facilitates PD-1 Checkpoint Therapy in Human Pancreatic Cancer
- Author
-
Venu G. Pillarisetty, Harlan Robins, Robert H. Pierce, Ian Nicholas Crispe, Raymond Yeung, Teresa S. Kim, Seth M. Pollack, Sara K. Daniel, James O. Park, Marissa Vignali, Arezou Abbasi, Kimberly S. Smythe, Florencia G. Jalikis, Kevin M. Sullivan, Xiuyun Jiang, and Yongwoo David Seo
- Abstract
Image cytometry methods overview
- Published
- 2023
13. Data from A Phase II Trial of Pembrolizumab and Vorinostat in Recurrent Metastatic Head and Neck Squamous Cell Carcinomas and Salivary Gland Cancer
- Author
-
Renato Martins, Keith Eaton, Laura Q.M. Chow, Christina S. Baik, Bernardo Goulart, Rafael Santana-Davila, Sylvia M. Lee, Venu G. Pillarisetty, Xiuyun Jiang, Jonathan R. Fromm, Jenna Voutsinas, Qian (Vicky) Wu, and Cristina P. Rodriguez
- Abstract
Purpose:This clinical trial combined pembrolizumab and vorinostat in recurrent/metastatic squamous cell carcinomas of the head and neck (HN), and salivary gland cancer (SGC).Patients and Methods:Patients with progressing incurable HN and SGC, Eastern Cooperative Oncology Group (ECOG) ≤1, no prior immunotherapy, RECIST1.1 measurable disease, and normal organ function were eligible. Pembrolizumab 200 mg was given intravenous every 21 days, and vorinostat 400 mg given orally 5 days on and 2 days off during each 21-day cycle. Primary endpoints were safety and objective response rates.Results:From November 2015 to August 2017, 25 patients with HN and 25 SGC were enrolled. Median age was 61 (range, 33–86) years, 39 (78%) were male, 21 (62%) were never smokers, and 27 (54%) had ECOG 0. In HN, 13 (52%) were p16+ oropharynx. Most common SGC histologies were adenoid cystic 12 (48%), acinic cell 3 (12%), and mucoepidermoid 3 (12%). Adverse events (AEs) in all patients were: 27 (54%) with grade ≥ 1 and 18 (36%) with grade ≥ 3. The most common AEs in all patients were renal insufficiency in seven, (14%), fatigue in six, (12%), and nausea in three (6%). There were three (12%) deaths on study. Responses in HN were complete response (CR) 0, partial response (PR) eight (32%), and stable disease (SD) five (20%). Efficacy in SGCs was CR 0, PR four (16%) in one lymphoepithelioma-like carcinoma, two acinic cell, one adenoid cystic, and SD 14 (56%). In the HN group, median follow-up (mFUP) was 12.6 months, median overall survival (mOS) was 12.6 months, and median progression-free survival (mPFS) was 4.5 months. In SGC, mFUP was 13.1 months, mOS was 14.0 months, and mPFS was 6.9 months.Conclusions:This combination demonstrated activity in HN, with fewer responses in SGC. Toxicities were higher than reported with pembrolizumab alone.
- Published
- 2023
14. Supplementary Tables 1-5 and Supplementary Figure Legends 1-3 from Predicting Survival in Patients Undergoing Resection for Locally Recurrent Retroperitoneal Sarcoma: A Study and Novel Nomogram from TARPSWG
- Author
-
Alessandro Gronchi, Mark Fairweather, Marco Fiore, Kenneth Cardona, Frits van Coevorden, Elisabetta Pennacchioli, Hans J. Gelderblom, John Mullen, Robert Canter, Sanjay Bagaria, Carol J. Swallow, Carolyn Nessim, Venu G. Pillarisetty, Antonino De Paoli, Eberhard Stoeckle, Vittorio Quagliuolo, Giovanni Grignani, Nita Ahuja, Ricardo Gonzalez, Dirk C. Strauss, Guy Lahat, Jean-Yves Blay, Piotr Rutkowski, Francesco Barretta, Rosalba Miceli, Dario Callegaro, and Chandrajit P. Raut
- Abstract
Supplementary Tables and Figure Legends
- Published
- 2023
15. Supplementary Data from A Phase II Trial of Pembrolizumab and Vorinostat in Recurrent Metastatic Head and Neck Squamous Cell Carcinomas and Salivary Gland Cancer
- Author
-
Renato Martins, Keith Eaton, Laura Q.M. Chow, Christina S. Baik, Bernardo Goulart, Rafael Santana-Davila, Sylvia M. Lee, Venu G. Pillarisetty, Xiuyun Jiang, Jonathan R. Fromm, Jenna Voutsinas, Qian (Vicky) Wu, and Cristina P. Rodriguez
- Abstract
Supplement Figure 1: Synergistic cytotoxic effect of pembrolizumab combined with vorinostat in HNSCC tissue slice culture. HNSCC slices (250 ï�m) were cultured and treated with 20 ï�g/ml of isotype control antibody IgG4 or pembrolizumab, with or without vorinostat (20 ï�M, 100 ï�M) for 6 days. Graph shows the percentage of cleaved-caspase-3+ cells. Unpaired student's tests, P-values are as follows: *p < 0.05, **P < 0.01, ***P< 0.001.
- Published
- 2023
16. Supplementary Figure 2 from Predicting Survival in Patients Undergoing Resection for Locally Recurrent Retroperitoneal Sarcoma: A Study and Novel Nomogram from TARPSWG
- Author
-
Alessandro Gronchi, Mark Fairweather, Marco Fiore, Kenneth Cardona, Frits van Coevorden, Elisabetta Pennacchioli, Hans J. Gelderblom, John Mullen, Robert Canter, Sanjay Bagaria, Carol J. Swallow, Carolyn Nessim, Venu G. Pillarisetty, Antonino De Paoli, Eberhard Stoeckle, Vittorio Quagliuolo, Giovanni Grignani, Nita Ahuja, Ricardo Gonzalez, Dirk C. Strauss, Guy Lahat, Jean-Yves Blay, Piotr Rutkowski, Francesco Barretta, Rosalba Miceli, Dario Callegaro, and Chandrajit P. Raut
- Abstract
Supplementary Figure 2
- Published
- 2023
17. Supplementary Figure 5 from Mobilization of CD8+ T Cells via CXCR4 Blockade Facilitates PD-1 Checkpoint Therapy in Human Pancreatic Cancer
- Author
-
Venu G. Pillarisetty, Harlan Robins, Robert H. Pierce, Ian Nicholas Crispe, Raymond Yeung, Teresa S. Kim, Seth M. Pollack, Sara K. Daniel, James O. Park, Marissa Vignali, Arezou Abbasi, Kimberly S. Smythe, Florencia G. Jalikis, Kevin M. Sullivan, Xiuyun Jiang, and Yongwoo David Seo
- Abstract
Live microscopy analysis of CD8+ T cell distribution in slice cultures
- Published
- 2023
18. Supplementary Figure 1B from Predicting Survival in Patients Undergoing Resection for Locally Recurrent Retroperitoneal Sarcoma: A Study and Novel Nomogram from TARPSWG
- Author
-
Alessandro Gronchi, Mark Fairweather, Marco Fiore, Kenneth Cardona, Frits van Coevorden, Elisabetta Pennacchioli, Hans J. Gelderblom, John Mullen, Robert Canter, Sanjay Bagaria, Carol J. Swallow, Carolyn Nessim, Venu G. Pillarisetty, Antonino De Paoli, Eberhard Stoeckle, Vittorio Quagliuolo, Giovanni Grignani, Nita Ahuja, Ricardo Gonzalez, Dirk C. Strauss, Guy Lahat, Jean-Yves Blay, Piotr Rutkowski, Francesco Barretta, Rosalba Miceli, Dario Callegaro, and Chandrajit P. Raut
- Abstract
Supplementary Figure 1F
- Published
- 2023
19. Supplementary Figure 3 from Predicting Survival in Patients Undergoing Resection for Locally Recurrent Retroperitoneal Sarcoma: A Study and Novel Nomogram from TARPSWG
- Author
-
Alessandro Gronchi, Mark Fairweather, Marco Fiore, Kenneth Cardona, Frits van Coevorden, Elisabetta Pennacchioli, Hans J. Gelderblom, John Mullen, Robert Canter, Sanjay Bagaria, Carol J. Swallow, Carolyn Nessim, Venu G. Pillarisetty, Antonino De Paoli, Eberhard Stoeckle, Vittorio Quagliuolo, Giovanni Grignani, Nita Ahuja, Ricardo Gonzalez, Dirk C. Strauss, Guy Lahat, Jean-Yves Blay, Piotr Rutkowski, Francesco Barretta, Rosalba Miceli, Dario Callegaro, and Chandrajit P. Raut
- Abstract
Supplementary Figure 3
- Published
- 2023
20. Supplementary Figure 3 from Mobilization of CD8+ T Cells via CXCR4 Blockade Facilitates PD-1 Checkpoint Therapy in Human Pancreatic Cancer
- Author
-
Venu G. Pillarisetty, Harlan Robins, Robert H. Pierce, Ian Nicholas Crispe, Raymond Yeung, Teresa S. Kim, Seth M. Pollack, Sara K. Daniel, James O. Park, Marissa Vignali, Arezou Abbasi, Kimberly S. Smythe, Florencia G. Jalikis, Kevin M. Sullivan, Xiuyun Jiang, and Yongwoo David Seo
- Abstract
PDA tumor slice culture platform utilized for multimodal analysis before and after ex vivo immune modulation
- Published
- 2023
21. Supplementary Figure 2 from Mobilization of CD8+ T Cells via CXCR4 Blockade Facilitates PD-1 Checkpoint Therapy in Human Pancreatic Cancer
- Author
-
Venu G. Pillarisetty, Harlan Robins, Robert H. Pierce, Ian Nicholas Crispe, Raymond Yeung, Teresa S. Kim, Seth M. Pollack, Sara K. Daniel, James O. Park, Marissa Vignali, Arezou Abbasi, Kimberly S. Smythe, Florencia G. Jalikis, Kevin M. Sullivan, Xiuyun Jiang, and Yongwoo David Seo
- Abstract
TCR overlap analysis
- Published
- 2023
22. Data from Mobilization of CD8+ T Cells via CXCR4 Blockade Facilitates PD-1 Checkpoint Therapy in Human Pancreatic Cancer
- Author
-
Venu G. Pillarisetty, Harlan Robins, Robert H. Pierce, Ian Nicholas Crispe, Raymond Yeung, Teresa S. Kim, Seth M. Pollack, Sara K. Daniel, James O. Park, Marissa Vignali, Arezou Abbasi, Kimberly S. Smythe, Florencia G. Jalikis, Kevin M. Sullivan, Xiuyun Jiang, and Yongwoo David Seo
- Abstract
Purpose:Pancreatic ductal adenocarcinoma (PDA) is rarely cured, and single-agent immune checkpoint inhibition has not demonstrated clinical benefit despite the presence of large numbers of CD8+ T cells. We hypothesized that tumor-infiltrating CD8+ T cells harbor latent antitumor activity that can be reactivated using combination immunotherapy.Experimental Design:Preserved human PDA specimens were analyzed using multiplex IHC (mIHC) and T-cell receptor (TCR) sequencing. Fresh tumor was treated in organotypic slice culture to test the effects of combination PD-1 and CXCR4 blockade. Slices were analyzed using IHC, flow cytometry, and live fluorescent microscopy to assess tumor kill, in addition to T-cell expansion and mobilization.Results:mIHC demonstrated fewer CD8+ T cells in juxtatumoral stroma containing carcinoma cells than in stroma devoid of them. Using TCR sequencing, we found clonal expansion in each tumor; high-frequency clones had multiple DNA rearrangements coding for the same amino acid binding sequence, which suggests response to common tumor antigens. Treatment of fresh human PDA slices with combination PD-1 and CXCR4 blockade led to increased tumor cell death concomitant with lymphocyte expansion. Live microscopy after combination therapy demonstrated CD8+ T-cell migration into the juxtatumoral compartment and rapid increase in tumor cell apoptosis.Conclusions:Endogenous tumor-reactive T cells are present within the human PDA tumor microenvironment and can be reactivated by combined blockade of PD-1 and CXCR4. This provides a new basis for the rational selection of combination immunotherapy for PDA.See related commentary by Medina and Miller, p. 3747
- Published
- 2023
23. Supplementary Table 1 from Mobilization of CD8+ T Cells via CXCR4 Blockade Facilitates PD-1 Checkpoint Therapy in Human Pancreatic Cancer
- Author
-
Venu G. Pillarisetty, Harlan Robins, Robert H. Pierce, Ian Nicholas Crispe, Raymond Yeung, Teresa S. Kim, Seth M. Pollack, Sara K. Daniel, James O. Park, Marissa Vignali, Arezou Abbasi, Kimberly S. Smythe, Florencia G. Jalikis, Kevin M. Sullivan, Xiuyun Jiang, and Yongwoo David Seo
- Abstract
Patient demographic and tumor pathology
- Published
- 2023
24. Supplementary Figure 1 from Mobilization of CD8+ T Cells via CXCR4 Blockade Facilitates PD-1 Checkpoint Therapy in Human Pancreatic Cancer
- Author
-
Venu G. Pillarisetty, Harlan Robins, Robert H. Pierce, Ian Nicholas Crispe, Raymond Yeung, Teresa S. Kim, Seth M. Pollack, Sara K. Daniel, James O. Park, Marissa Vignali, Arezou Abbasi, Kimberly S. Smythe, Florencia G. Jalikis, Kevin M. Sullivan, Xiuyun Jiang, and Yongwoo David Seo
- Abstract
Image cytometry infiltration results by layer
- Published
- 2023
25. Data from Predicting Survival in Patients Undergoing Resection for Locally Recurrent Retroperitoneal Sarcoma: A Study and Novel Nomogram from TARPSWG
- Author
-
Alessandro Gronchi, Mark Fairweather, Marco Fiore, Kenneth Cardona, Frits van Coevorden, Elisabetta Pennacchioli, Hans J. Gelderblom, John Mullen, Robert Canter, Sanjay Bagaria, Carol J. Swallow, Carolyn Nessim, Venu G. Pillarisetty, Antonino De Paoli, Eberhard Stoeckle, Vittorio Quagliuolo, Giovanni Grignani, Nita Ahuja, Ricardo Gonzalez, Dirk C. Strauss, Guy Lahat, Jean-Yves Blay, Piotr Rutkowski, Francesco Barretta, Rosalba Miceli, Dario Callegaro, and Chandrajit P. Raut
- Abstract
Purpose:The role of surgery for first relapse locally recurrent retroperitoneal sarcoma (RPS-LR1) is uncertain. We report outcomes of the largest RPS-LR1 series and propose a new prognostic nomogram.Experimental Design:Patients with consecutive RPS-LR1 without distant metastases who underwent resection at 22 centers (2002–2011) were included. Endpoints were disease-free and overall survival (DFS, OS) and crude-cumulative-incidence (CCI) of local/distant recurrence from second surgery. Nomograms predicting DFS and OS from second surgery were developed and validated (calibration plots); discrimination was assessed (Harrell C index).Results:Of 684 patients identified, full prognostic variable data were available for 602. Initial surgery for primary RPS was performed at our institutions in 188 patients (31%) and elsewhere in 414 (69%). At a median follow-up of 119 months [Interquartile range (IQR), 80–169] from initial surgery and 75 months (IQR 50–105) from second surgery, 6-year DFS and OS were 19.2% [95% confidence interval (CI), 16.0–23.0%] and 54.1% (95% CI, 49.8–58.8%), respectively. Recurrence patterns and survival probability were histology-specific, with liposarcoma subtypes having the highest 6-year CCI of second local recurrence (LR, 60.2%–70.9%) and leiomyosarcoma (LMS) having higher 6-year CCI of distant metastasis (DM, 36.3%). Nomograms included age at second surgery, multifocality, grade, completeness of second surgery, histology, chemotherapy/radiotherapy at first surgery, and number of organs resected at first surgery. OS and DFS nomograms showed good calibration and discriminative ability (C index 0.70 and 0.67, respectively).Conclusions:We developed nomograms to predict DFS and OS for patients undergoing RPS-LR1 resection. Nomograms provide individualized, disease-relevant estimations of survival for RPS-LR1 patients and assist in clinical decisions.
- Published
- 2023
26. Supplementary Figure 4 from Mobilization of CD8+ T Cells via CXCR4 Blockade Facilitates PD-1 Checkpoint Therapy in Human Pancreatic Cancer
- Author
-
Venu G. Pillarisetty, Harlan Robins, Robert H. Pierce, Ian Nicholas Crispe, Raymond Yeung, Teresa S. Kim, Seth M. Pollack, Sara K. Daniel, James O. Park, Marissa Vignali, Arezou Abbasi, Kimberly S. Smythe, Florencia G. Jalikis, Kevin M. Sullivan, Xiuyun Jiang, and Yongwoo David Seo
- Abstract
Combination immunotherapy tumor slice culture experiment replicates
- Published
- 2023
27. Long-lived pancreatic ductal adenocarcinoma slice cultures enable precise study of the immune microenvironment
- Author
-
Xiuyun Jiang, Y. David Seo, Jae Hyuck Chang, Andrew Coveler, Eslam N. Nigjeh, Sheng Pan, Florencia Jalikis, Raymond S. Yeung, Ian N. Crispe, and Venu G. Pillarisetty
- Subjects
immune cells ,model ,pancreatic ductal adenocarcinoma ,pda ,slice culture ,Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Pancreatic ductal adenocarcinoma (PDA) remains a deadly disease that is rarely cured, despite many recent successes with immunotherapy for other malignancies. As the human disease is heavily infiltrated by effector T cells, we postulated that accurately modeling the PDA immune microenvironment would allow us to study mechanisms of immunosuppression that could be overcome for therapeutic benefit. Using viable precision-cut slices from fresh PDA, we developed an organotypic culture system for this purpose. We confirmed that cultured slices maintain their baseline morphology, surface area, and microenvironment after at least 6 d in culture, and demonstrated slice survival by MTT assay and by immunohistochemistry staining with Ki-67 and cleaved-Caspase-3 antibodies. Immune cells, including T cells (CD3+, CD8+, and FOXP3+) and macrophages (CD68+, CD163+ and HLA-DR+), as well as stromal myofibroblasts (αSMA+) were present throughout the culture period. Global profiling of the PDA proteome before and after 6 d slice culture indicated that the majority of the immunological proteins identified remain stable during the culture process. Cytotoxic effects of drug treatment (staurosporine, STS and cycloheximide, CHX) on PDA slices culture confirmed that this system can be used to assess functional response and cell survival following drug treatment in both a treatment time- and dose-dependent manner. Using multicolor immunofluorescence, we stained live slices for both cancer cells (EpCAM+) and immune cells (CD11b+ and CD8+). Finally, we confirmed that autologous CFSE-labeled splenocytes readily migrate into co-cultured tumor slices. Thus, our present study demonstrates the potential to use tumor slice cultures to study the immune microenvironment of PDA.
- Published
- 2017
- Full Text
- View/download PDF
28. IWATE criteria are associated with perioperative outcomes in robotic hepatectomy: a retrospective review of 225 resections
- Author
-
Kevin M. Sullivan, Alex W. Lois, Lindsay K. Dickerson, Sara K. Daniel, Raymond S. Yeung, Jonathan G. Sham, Kyle S. Bilodeau, James O. Park, Jaqueline Valdez Gonzalez, Alan F. Utria, Kevin P. Labadie, Venu G. Pillarisetty, David J. Droullard, Kathryn E. McNevin, John Calhoun, and Yongwoo D. Seo
- Subjects
medicine.medical_specialty ,medicine.medical_treatment ,030230 surgery ,Article ,03 medical and health sciences ,Postoperative Complications ,0302 clinical medicine ,Robotic Surgical Procedures ,Minimally invasive surgery ,Internal medicine ,Hepatectomy ,Humans ,Medicine ,Robotic surgery ,Intrahepatic Cholangiocarcinoma ,Robotic hepatectomy ,Retrospective Studies ,business.industry ,Liver Neoplasms ,Confounding ,Perioperative ,Length of Stay ,Hepatology ,medicine.disease ,Surgery ,Bile Ducts, Intrahepatic ,Bile Duct Neoplasms ,IWATE criteria ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Laparoscopy ,business ,Abdominal surgery - Abstract
Background Robotic hepatectomy (RH) is increasingly utilized for minor and major liver resections. The IWATE criteria were developed to classify minimally invasive liver resections by difficulty. The objective of this study was to apply the IWATE criteria in RH and to describe perioperative and oncologic outcomes of RH over the last decade at our institution. Methods Perioperative and oncologic outcomes of patients who underwent RH between 2011 and 2019 were retrospectively collected. The difficulty level of each operation was assessed using the IWATE criteria, and outcomes were compared at each level. Univariate linear regression was performed to characterize the relationship between IWATE criteria and perioperative outcomes (OR time, EBL, and LOS), and a multivariable model was also developed to address potential confounding by patient characteristics (age, sex, BMI, prior abdominal surgery, ASA class, and simultaneous non-hepatectomy operation). Results Two hundred and twenty-five RH were performed. Median IWATE criteria for RH were 6 (IQR 5–9), with low, intermediate, advanced, and expert resections accounting for 23% (n = 51), 34% (n = 77), 32% (n = 72), and 11% (n = 25) of resections, respectively. The majority of resections were parenchymal-sparing approaches, including anatomic segmentectomies and non-anatomic partial resections. 30-day complication rate was 14%, conversion to open surgery occurred in 9 patients (4%), and there were no deaths within 30 days postoperatively. In the univariate linear regression analysis, IWATE criteria were positively associated with OR time, EBL, and LOS. In the multivariable model, IWATE criteria were independently associated with greater OR time, EBL, and LOS. Two-year overall survival for hepatocellular carcinoma and intrahepatic cholangiocarcinoma was 94% and 50%, respectively. Conclusion In conclusion, the IWATE criteria are associated with surgical outcomes after RH. This series highlights the utility of RH for difficult hepatic resections, particularly parenchymal-sparing resections in the posterosuperior sector, extending the indication of minimally invasive hepatectomy in experienced hands and potentially offering select patients an alternative to open hepatectomy or other less definitive liver-directed treatment options.
- Published
- 2021
29. Key chemokines direct migration of immune cells in solid tumors
- Author
-
Karan Kohli, Venu G. Pillarisetty, and Teresa S. Kim
- Subjects
0301 basic medicine ,Cancer Research ,Chemokine ,animal diseases ,chemical and pharmacologic phenomena ,Context (language use) ,Review Article ,Biology ,03 medical and health sciences ,Chemokine receptor ,0302 clinical medicine ,Immune system ,Cell Movement ,Neoplasms ,Tumor Microenvironment ,Humans ,Molecular Biology ,Immune cell infiltration ,Cancer immunology ,Tumor microenvironment ,Effector ,biochemical phenomena, metabolism, and nutrition ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,bacteria ,Tumour immunology ,Molecular Medicine ,Chemokines - Abstract
Immune cell infiltration into solid tumors, their movement within the tumor microenvironment (TME), and interaction with other immune cells are controlled by their directed migration towards gradients of chemokines. Dysregulated chemokine signaling in TME favors the growth of tumors, exclusion of effector immune cells, and abundance of immunosuppressive cells. Key chemokines directing the migration of immune cells into tumor tissue have been identified. In this review, we discuss well-studied chemokine receptors that regulate migration of effector and immunosuppressive immune cells in the context of cancer immunology. We discuss preclinical models that have described the role of respective chemokine receptors in immune cell migration into TME and review preclinical and clinical studies that target chemokine signaling as standalone or combination therapies.
- Published
- 2021
30. A Phase II Trial of Pembrolizumab and Vorinostat in Recurrent Metastatic Head and Neck Squamous Cell Carcinomas and Salivary Gland Cancer
- Author
-
Jenna M. Voutsinas, Laura Q.M. Chow, Renato G. Martins, Venu G. Pillarisetty, Keith D. Eaton, Jonathan R. Fromm, Christina S. Baik, Rafael Santana-Davila, Bernardo H. L. Goulart, Sylvia Lee, Xiuyun Jiang, Cristina P. Rodriguez, and Qian Vicky Wu
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Nausea ,Pembrolizumab ,Antibodies, Monoclonal, Humanized ,Adenoid ,Gastroenterology ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Carcinoma ,medicine ,Humans ,Neoplasm Metastasis ,Adverse effect ,Survival rate ,Aged ,Aged, 80 and over ,Vorinostat ,Squamous Cell Carcinoma of Head and Neck ,business.industry ,Middle Aged ,Salivary Gland Neoplasms ,medicine.disease ,Survival Rate ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Drug Resistance, Neoplasm ,Head and Neck Neoplasms ,Salivary gland cancer ,030220 oncology & carcinogenesis ,HN group ,Female ,Neoplasm Recurrence, Local ,medicine.symptom ,business - Abstract
Purpose: This clinical trial combined pembrolizumab and vorinostat in recurrent/metastatic squamous cell carcinomas of the head and neck (HN), and salivary gland cancer (SGC). Patients and Methods: Patients with progressing incurable HN and SGC, Eastern Cooperative Oncology Group (ECOG) ≤1, no prior immunotherapy, RECIST1.1 measurable disease, and normal organ function were eligible. Pembrolizumab 200 mg was given intravenous every 21 days, and vorinostat 400 mg given orally 5 days on and 2 days off during each 21-day cycle. Primary endpoints were safety and objective response rates. Results: From November 2015 to August 2017, 25 patients with HN and 25 SGC were enrolled. Median age was 61 (range, 33–86) years, 39 (78%) were male, 21 (62%) were never smokers, and 27 (54%) had ECOG 0. In HN, 13 (52%) were p16+ oropharynx. Most common SGC histologies were adenoid cystic 12 (48%), acinic cell 3 (12%), and mucoepidermoid 3 (12%). Adverse events (AEs) in all patients were: 27 (54%) with grade ≥ 1 and 18 (36%) with grade ≥ 3. The most common AEs in all patients were renal insufficiency in seven, (14%), fatigue in six, (12%), and nausea in three (6%). There were three (12%) deaths on study. Responses in HN were complete response (CR) 0, partial response (PR) eight (32%), and stable disease (SD) five (20%). Efficacy in SGCs was CR 0, PR four (16%) in one lymphoepithelioma-like carcinoma, two acinic cell, one adenoid cystic, and SD 14 (56%). In the HN group, median follow-up (mFUP) was 12.6 months, median overall survival (mOS) was 12.6 months, and median progression-free survival (mPFS) was 4.5 months. In SGC, mFUP was 13.1 months, mOS was 14.0 months, and mPFS was 6.9 months. Conclusions: This combination demonstrated activity in HN, with fewer responses in SGC. Toxicities were higher than reported with pembrolizumab alone.
- Published
- 2020
31. DNA Damage Repair Defects and Survival Outcomes for Patients With Resected Pancreatic Ductal Adenocarcinoma
- Author
-
Marc R. Radke, Elena G. Chiorean, Deepti M. Reddi, Venu G. Pillarisetty, Kelsey Baker, Elizabeth M. Swisher, Amy Chang, Kit Man Wong, Andrew L. Coveler, David Bing Zhen, and Mary W. Redman
- Subjects
Time Factors ,Pancreatic ductal adenocarcinoma ,Hepatology ,business.industry ,Endocrinology, Diabetes and Metabolism ,Recombinational DNA Repair ,DNA Damage Repair ,Article ,Pancreatic Neoplasms ,Pancreatectomy ,Treatment Outcome ,Endocrinology ,Text mining ,Mutation ,Biomarkers, Tumor ,Internal Medicine ,Cancer research ,Humans ,Medicine ,business ,Carcinoma, Pancreatic Ductal ,DNA Damage ,Retrospective Studies - Published
- 2021
32. Impact of enhanced recovery protocols after pancreatoduodenectomy: meta-analysis
- Author
-
Christoph Kuemmerli, Christoph Tschuor, Meidai Kasai, Adnan A Alseidi, Gianpaolo Balzano, Stefan Bouwense, Marco Braga, Mariëlle Coolsen, Sara K Daniel, Christos Dervenis, Massimo Falconi, Dae Wook Hwang, Daniel J Kagedan, Song Cheol Kim, Harish Lavu, Tingbo Liang, Daniel Nussbaum, Stefano Partelli, Michael J Passeri, Nicolò Pecorelli, Sastha Ahanatha Pillai, Venu G Pillarisetty, Michael J Pucci, Wei Su, Robert P Sutcliffe, Bobby Tingstedt, Marion van der Kolk, Dionisios Vrochides, Alice Wei, Caroline Williamsson, Charles J Yeo, Sabino Zani, Efstratios Zouros, Mohammed Abu Hilal, Kuemmerli, Christoph, Tschuor, Christoph, Kasai, Meidai, Alseidi, Adnan A, Balzano, Gianpaolo, Bouwense, Stefan, Braga, Marco, Coolsen, Mariëlle, Daniel, Sara K, Dervenis, Christo, Falconi, Massimo, Hwang, Dae Wook, Kagedan, Daniel J, Kim, Song Cheol, Lavu, Harish, Liang, Tingbo, Nussbaum, Daniel, Partelli, Stefano, Passeri, Michael J, Pecorelli, Nicolò, Pillai, Sastha Ahanatha, Pillarisetty, Venu G, Pucci, Michael J, Su, Wei, Sutcliffe, Robert P, Tingstedt, Bobby, van der Kolk, Marion, Vrochides, Dionisio, Wei, Alice, Williamsson, Caroline, Yeo, Charles J, Zani, Sabino, Zouros, Efstratio, and Abu Hilal, Mohammed
- Subjects
COMPLICATIONS ,Postoperative Complications/prevention & control ,FAST-TRACK SURGERY ,LENGTH-OF-STAY ,Recovery of Function ,Length of Stay ,PANCREATIC SURGERY ,Patient Readmission ,Pancreaticoduodenectomy ,PATHWAY ,COLORECTAL SURGERY ,Postoperative Complications ,MANAGEMENT ,IMPLEMENTATION ,PROGRAM ,Humans ,Surgery ,Pancreaticoduodenectomy/adverse effects ,PERIOPERATIVE CARE ,Enhanced Recovery After Surgery - Abstract
Background This individual-patient data meta-analysis investigated the effects of enhanced recovery after surgery (ERAS) protocols compared with conventional care on postoperative outcomes in patients undergoing pancreatoduodenectomy.Methods The Cochrane Library, MEDLINE, Embase, Scopus, and Web of Science were searched systematically for articles reporting outcomes of ERAS after pancreatoduodenectomy published up to August 2020. Comparative studies were included. Main outcomes were postoperative functional recovery elements, postoperative morbidity, duration of hospital stay, and readmission.Results Individual-patient data were obtained from 17 of 31 eligible studies comprising 3108 patients. Time to liquid (mean difference (MD) -3.23 (95 per cent c.i. -4.62 to -1.85) days; P < 0.001) and solid (-3.84 (-5.09 to -2.60) days; P < 0.001) intake, time to passage of first stool (MD -1.38 (-1.82 to -0.94) days; P < 0.001) and time to removal of the nasogastric tube (3.03 (-4.87 to -1.18) days; P = 0.001) were reduced with ERAS. ERAS was associated with lower overall morbidity (risk difference (RD) -0.04, 95 per cent c.i. -0.08 to -0.01; P = 0.015), less delayed gastric emptying (RD -0.11, -0.22 to -0.01; P = 0.039) and a shorter duration of hospital stay (MD -2.33 (-2.98 to -1.69) days; P < 0.001) without a higher readmission rate.Conclusion ERAS improved postoperative outcome after pancreatoduodenectomy. Implementation should be encouraged.Lay SummaryEnhanced recovery protocols consist of interdisciplinary interventions aimed at standardizing care and reducing the impact of surgical stress. They often include a short period of preoperative fasting during the night before surgery, early removal of lines and surgical drains, early food intake and mobilization out of bed on the day of surgery. This study gives a summary of reports assessing such care protocols in patients undergoing pancreatic head surgery, and assesses the impact of these protocols on functional recovery in an analysis of individual-patient data. The study revealed the true benefits of enhanced recovery protocols, including shorter time to food intake, earlier bowel activity, fewer complications after surgery, and a shorter hospital stay compared with conventional care.
- Published
- 2021
33. Blockade of interleukin 10 potentiates antitumour immune function in human colorectal cancer liver metastases
- Author
-
Kevin M Sullivan, Xiuyun Jiang, Prajna Guha, Christopher Lausted, Jason A Carter, Cynthia Hsu, Kevin P Labadie, Karan Kohli, Heidi L Kenerson, Sara K Daniel, Xiaowei Yan, Changting Meng, Arezou Abbasi, Marina Chan, Y David Seo, James O Park, Ian Nicholas Crispe, Raymond S Yeung, Teresa S Kim, Taranjit S Gujral, Qiang Tian, Steven C Katz, and Venu G Pillarisetty
- Subjects
Gastroenterology - Abstract
ObjectiveProgrammed cell death protein 1 (PD-1) checkpoint inhibition and adoptive cellular therapy have had limited success in patients with microsatellite stable colorectal cancer liver metastases (CRLM). We sought to evaluate the effect of interleukin 10 (IL-10) blockade on endogenous T cell and chimeric antigen receptor T (CAR-T) cell antitumour function in CRLM slice cultures.DesignWe created organotypic slice cultures from human CRLM (n=38 patients’ tumours) and tested the antitumour effects of a neutralising antibody against IL-10 (αIL-10) both alone as treatment and in combination with exogenously administered carcinoembryonic antigen (CEA)-specific CAR-T cells. We evaluated slice cultures with single and multiplex immunohistochemistry, in situ hybridisation, single-cell RNA sequencing, reverse-phase protein arrays and time-lapse fluorescent microscopy.ResultsαIL-10 generated a 1.8-fold increase in T cell-mediated carcinoma cell death in human CRLM slice cultures. αIL-10 significantly increased proportions of CD8+T cells without exhaustion transcription changes, and increased human leukocyte antigen - DR isotype (HLA-DR) expression of macrophages. The antitumour effects of αIL-10 were reversed by major histocompatibility complex class I or II (MHC-I or MHC-II) blockade, confirming the essential role of antigen presenting cells. Interrupting IL-10 signalling also rescued murine CAR-T cell proliferation and cytotoxicity from myeloid cell-mediated immunosuppression. In human CRLM slices, αIL-10 increased CEA-specific CAR-T cell activation and CAR-T cell-mediated cytotoxicity, with nearly 70% carcinoma cell apoptosis across multiple human tumours. Pretreatment with an IL-10 receptor blocking antibody also potentiated CAR-T function.ConclusionNeutralising the effects of IL-10 in human CRLM has therapeutic potential as a stand-alone treatment and to augment the function of adoptively transferred CAR-T cells.
- Published
- 2021
34. Neuroendocrine and Adrenal Tumors, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology
- Author
-
Arash Kardan, Thomas J. Giordano, Thorvardur R. Halfdanarson, Jennifer A. Chan, Lawrence S. Blaszkowsky, Christopher H. Lieu, Pamela Brock, Anthony P. Heaney, Boris W. Kuvshinoff, Satya Das, Whitney S. Goldner, Al B. Benson, Cindy Hochstetler, Beth Lynn, Heloisa P. Soares, Craig R. Sussman, Fouad Kandeel, Jin He, Daniel M. Halperin, Emily K. Bergsland, Jonathan R. Strosberg, Nataliya Uboha, Sajid A. Khan, Terence Z. Wong, Kimberly A. Miller, Michael C. Soulen, Martin J. Heslin, Venu G. Pillarisetty, Manisha H. Shah, Paxton V. Dickson, Nikolaos A. Trikalinos, Namrata Vijayvergia, Shagufta Shaheen, Diane Lauren Reidy, Sarimar N Agosto Salgado, and Paul T. Fanta
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Adrenal Gland Neoplasms ,Neuroendocrine tumors ,Medical Oncology ,Surgical methods ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Genetic risk ,Stage (cooking) ,Multiple endocrine neoplasia ,Adrenal tumors ,business.industry ,Adrenal gland ,Poorly differentiated ,medicine.disease ,Neuroendocrine Tumors ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,business - Abstract
The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Neuroendocrine and Adrenal Gland Tumors focus on the diagnosis, treatment, and management of patients with neuroendocrine tumors (NETs), adrenal tumors, pheochromocytomas, paragangliomas, and multiple endocrine neoplasia. NETs are generally subclassified by site of origin, stage, and histologic characteristics. Appropriate diagnosis and treatment of NETs often involves collaboration between specialists in multiple disciplines, using specific biochemical, radiologic, and surgical methods. Specialists include pathologists, endocrinologists, radiologists (including nuclear medicine specialists), and medical, radiation, and surgical oncologists. These guidelines discuss the diagnosis and management of both sporadic and hereditary neuroendocrine and adrenal tumors and are intended to assist with clinical decision-making. This article is focused on the 2021 NCCN Guidelines principles of genetic risk assessment and counseling and recommendations for well-differentiated grade 3 NETs, poorly differentiated neuroendocrine carcinomas, adrenal tumors, pheochromocytomas, and paragangliomas.
- Published
- 2021
35. Epidermoid Cyst within an Intrapancreatic Accessory Spleen Mimicking a Pancreatic Cystic Neoplasm
- Author
-
Venu G Pillarisetty, Arezou Abbasi, Florencia G Jalikis, and Lisa K Koch
- Subjects
education.field_of_study ,Pathology ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Splenectomy ,Population ,Epidermoid cyst ,Accessory spleen ,Malignancy ,medicine.disease ,Pancreatic cystic neoplasm ,medicine ,Differential diagnosis ,business ,Complication ,education - Abstract
Although intra-abdominal accessory spleens are commonly found in 10-30% of the general population, epidermoid cyst within an intrapancreatic accessory spleen (ECIPAS) is an extremely rare entity and is often misdiagnosed preoperatively as a cystic malignancy. We present the case of a 51-year-old man who was referred to our clinic because of an incidentally found pancreatic tail cystic mass. Due to CA 19-9 level over 2000 and high suspicion of malignancy, the patient underwent distal pancreatectomy with splenectomy. Histopathological analysis revealed a squamous epithelial lining with splenic parenchyma within the cyst wall, which was consistent with ECIPAS. The patient was discharged on postoperative day 4 without any complication. As this disease may mimic malignancy with no characteristic features in preoperative imaging, it should be considered in the differential diagnosis of pancreatic cystic lesions.
- Published
- 2020
36. Systemic Interferon-γ Increases MHC Class I Expression and T-cell Infiltration in Cold Tumors: Results of a Phase 0 Clinical Trial
- Author
-
Seth M. Pollack, Stanley R. Riddell, Venu G. Pillarisetty, Qianchuan He, Cassian Yee, Robin L. Jones, Robert H. Pierce, R. Graeme Black, Lee D. Cranmer, Shihong Zhang, Brian A. Van Tine, Sydney Spadinger, Lu Yao, and Karan Kohli
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,Biopsy ,T-Lymphocytes ,medicine.medical_treatment ,Immunology ,Major histocompatibility complex ,Article ,Immunophenotyping ,Interferon-gamma ,Sarcoma, Synovial ,Young Adult ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Antigen ,Cancer immunotherapy ,Antigens, Neoplasm ,MHC class I ,medicine ,Humans ,Aged ,Tumor microenvironment ,biology ,business.industry ,Histocompatibility Antigens Class I ,Immunotherapy ,Middle Aged ,Liposarcoma, Myxoid ,Tumor antigen ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Cytokines ,Female ,business ,Biomarkers - Abstract
Interferon-γ (IFNγ) has been studied as a cancer treatment with limited evidence of clinical benefit. However, it could play a role in cancer immunotherapy combination treatments. Despite high expression of immunogenic cancer–testis antigens, synovial sarcoma (SS) and myxoid/round cell liposarcoma (MRCL) have a cold tumor microenvironment (TME), with few infiltrating T cells and low expression of major histocompatibility complex class I (MHC-I). We hypothesized that IFNγ treatment could drive inflammation in a cold TME, facilitating further immunotherapy. We conducted a phase 0 clinical trial treating 8 SS or MRCL patients with weekly systemic IFNγ. We performed pre- and posttreatment biopsies. IFNγ changed the SS and MRCL TME, inducing tumor-surface MHC-I expression and significant T-cell infiltration (P < 0.05). Gene-expression analysis suggested increased tumor antigen presentation and less exhausted phenotypes of the tumor-infiltrating T cells. Newly emergent antigen-specific humoral and/or T-cell responses were found in 3 of 7 evaluable patients. However, increased expression of PD-L1 was observed on tumor-infiltrating myeloid cells and in some cases tumor cells. These findings suggest that systemic IFNγ used to convert SS and MRCL into “hot” tumors will work in concert with anti–PD-1 therapy to provide patient benefit.
- Published
- 2019
37. Hypoxia as a barrier to immunotherapy in pancreatic adenocarcinoma
- Author
-
Kevin M. Sullivan, Sara K. Daniel, Venu G. Pillarisetty, and Kevin P. Labadie
- Subjects
0301 basic medicine ,medicine.medical_treatment ,T cell ,Medicine (miscellaneous) ,Review ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Cytotoxic T cell ,Hypoxia ,Tumor microenvironment ,lcsh:R5-920 ,Solid tumor ,Chemistry ,Immunotherapy ,Pancreatic cancer ,030104 developmental biology ,Cytokine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Myeloid-derived Suppressor Cell ,Molecular Medicine ,lcsh:Medicine (General) - Abstract
Pancreatic ductal adenocarcinoma (PDA) is a lethal disease with limited response to cytotoxic chemoradiotherapy, as well as newer immunotherapies. The PDA tumor microenvironment contains infiltrating immune cells including cytotoxic T cells; however, there is an overall immunosuppressive milieu. Hypoxia is a known element of the solid tumor microenvironment and may promote tumor survival. Through various mechanisms including, but not limited to, those mediated by HIF-1α, hypoxia also leads to increased tumor proliferation and metabolic changes. Furthermore, epithelial to mesenchymal transition is promoted through several pathways, including NOTCH and c-MET, regulated by hypoxia. Hypoxia-promoted changes also contribute to the immunosuppressive phenotype seen in many different cell types within the microenvironment and thereby may inhibit an effective immune system response to PDA. Pancreatic stellate cells (PSCs) and myofibroblasts appear to contribute to the recruitment of myeloid derived suppressor cells (MDSCs) and B cells in PDA via cytokines increased due to hypoxia. PSCs also increase collagen secretion in response to HIF-1α, which promotes a fibrotic stroma that alters T cell homing and migration. In hypoxic environments, B cells contribute to cytotoxic T cell exhaustion and produce chemokines to attract more immunosuppressive regulatory T cells. MDSCs inhibit T cell metabolism by hoarding key amino acids, modulate T cell homing by cleaving L-selectin, and prevent T cell activation by increasing PD-L1 expression. Immunosuppressive M2 phenotype macrophages promote T cell anergy via increased nitric oxide (NO) and decreased arginine in hypoxia. Increased numbers of regulatory T cells are seen in hypoxia which prevent effector T cell activation through cytokine production and increased CTLA-4. Effective immunotherapy for pancreatic adenocarcinoma and other solid tumors will need to help counteract the immunosuppressive nature of hypoxia-induced changes in the tumor microenvironment. Promising studies will look at combination therapies involving checkpoint inhibitors, chemokine inhibitors, and possible targeting of hypoxia. While no model is perfect, assuring that models incorporate the effects of hypoxia on cancer cells, stromal cells, and effector immune cells will be crucial in developing successful therapies.
- Published
- 2019
38. Hypoxia-inducible lentiviral gene expression in engineered human macrophages
- Author
-
Harrison K Chinn, Jennifer L Gardell, Lisa R Matsumoto, Kevin P Labadie, Tara N Mihailovic, Nicole A P Lieberman, Amira Davis, Venu G Pillarisetty, and Courtney A Crane
- Subjects
Pharmacology ,Cancer Research ,Macrophages ,Lentivirus ,Immunology ,Gene Expression ,Cell Hypoxia ,Mice ,Oncology ,Tumor Microenvironment ,Animals ,Cytokines ,Humans ,Molecular Medicine ,Immunology and Allergy - Abstract
BackgroundHuman immune cells, including monocyte-derived macrophages, can be engineered to deliver proinflammatory cytokines, bispecific antibodies, and chimeric antigen receptors to support immune responses in different disease settings. When gene expression is regulated by constitutively active promoters, lentiviral payload gene expression is unregulated, and can result in potentially toxic quantities of proteins. Regulated delivery of lentivirally encoded proteins may allow localized or conditional therapeutic protein expression to support safe delivery of adoptively transferred, genetically modified cells with reduced capacity for systemic toxicities.MethodsIn this study, we engineered human macrophages to express genes regulated by hypoxia responsive elements included in the lentiviral promoter region to drive conditional lentiviral gene expression only under hypoxic conditions. We tested transduced macrophages cultured in hypoxic conditions for the transient induced expression of reporter genes and the secreted cytokine, interleukin-12. Expression of hypoxia-regulated genes was investigated both transcriptionally and translationally, and in the presence of human tumor cells in a slice culture system. Finally, hypoxia-regulated gene expression was evaluated in a subcutaneous humanized-mouse cancer model.ResultsEngineered macrophages were shown to conditionally and tranisently express lentivirally encoded gene protein products, including IL-12 in hypoxic conditions in vitro. On return to normoxic conditions, lentiviral payload expression returned to basal levels. Reporter genes under the control of hypoxia response elements were upregulated under hypoxic conditions in the presence of human colorectal carcinoma cells and in the hypoxic xenograft model of glioblastoma, suggesting utility for systemic engineered cell delivery capable of localized gene delivery in cancer.ConclusionsMacrophages engineered to express hypoxia-regulated payloads have the potential to be administered systemically and conditionally express proteins in tissues with hypoxic conditions. In contrast to immune cells that function or survive poorly in hypoxic conditions, macrophages maintain a proinflammatory phenotype that may support continued gene and protein expression when regulated by conditional hypoxia responsive elements and naturally traffic to hypoxic microenvironments, making them ideal vehicles for therapeutic payloads to hypoxic tissues, such as solid tumors. With the ability to fine-tune delivery of potent proteins in response to endogenous microenvironments, macrophage-based cellular therapies may therefore be designed for different disease settings.
- Published
- 2022
39. Neoantigen-specific CD4+ T cells in human melanoma have diverse differentiation states and correlate with CD8+ T cell, macrophage, and B cell function
- Author
-
Joshua R. Veatch, Sylvia M. Lee, Carolyn Shasha, Naina Singhi, Julia L. Szeto, Ata S. Moshiri, Teresa S. Kim, Kimberly Smythe, Paul Kong, Matthew Fitzgibbon, Brenda Jesernig, Shailender Bhatia, Scott S. Tykodi, Evan T. Hall, David R. Byrd, John A. Thompson, Venu G. Pillarisetty, Thomas Duhen, A. McGarry Houghton, Evan Newell, Raphael Gottardo, and Stanley R. Riddell
- Subjects
CD4-Positive T-Lymphocytes ,Mice ,Cancer Research ,Oncology ,Antigens, Neoplasm ,Macrophages ,Tumor Microenvironment ,Animals ,Humans ,CD8-Positive T-Lymphocytes ,Melanoma ,Article - Abstract
CD4(+) T cells that recognize tumor antigens are required for immune checkpoint inhibitor efficacy in murine models but their contributions in human cancer are unclear. We used single cell RNA sequencing and T cell receptor sequences to identify signatures and functional correlates of tumor specific CD4(+) T cells infiltrating human melanoma. Conventional CD4(+) T cells that recognize tumor neoantigens express CXCL13 and are subdivided into clusters expressing memory and T follicular helper markers, and those expressing cytolytic markers, inhibitory receptors, and IFN-γ. The frequency of CXCL13(+) CD4(+) T cells in the tumor correlated with the transcriptional states of CD8(+) T cells and macrophages, maturation of B cells, and patient survival. Similar correlations were observed in a breast cancer cohort. These results identify phenotypes and functional correlates of tumor specific CD4(+) T cells in melanoma and suggest the possibility of using such cells to modify the tumor microenvironment.
- Published
- 2022
40. Genetically engineered macrophages persist in solid tumors and locally deliver therapeutic proteins to activate immune responses
- Author
-
Shannon A Kreuser, Venu G. Pillarisetty, Robert H. Pierce, Nicole A P Lieberman, Courtney A. Crane, Michael C. Jensen, Stephanie Balcaitis, Kimberly S. Smythe, Lisa R Matsumoto, Kole DeGolier, Virginia Hoglund, Katherine J. Brempelis, Chibawanye I. Ene, Amira Davis, Kevin P. Labadie, Harrison Chinn, Richard G. Ellenbogen, Eric C. Holland, Jason K. Yokoyama, Brooke M Prieskorn, Kara White Moyes, Sara K. Daniel, Courtney M Cowan, and Jean S. Campbell
- Subjects
Cancer Research ,Chemokine ,medicine.medical_treatment ,Immunology ,cell engineering ,CD19 ,Mice ,Immune system ,Neoplasms ,medicine ,Tumor Microenvironment ,Immunology and Allergy ,Animals ,Humans ,RC254-282 ,Pharmacology ,Tumor microenvironment ,biology ,Immune Cell Therapies and Immune Cell Engineering ,Melanoma ,Macrophages ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunotherapy ,medicine.disease ,Chimeric antigen receptor ,Disease Models, Animal ,Oncology ,biology.protein ,Cancer research ,Molecular Medicine ,Genetic Engineering ,Ex vivo - Abstract
BackgroundThough currently approved immunotherapies, including chimeric antigen receptor T cells and checkpoint blockade antibodies, have been successfully used to treat hematological and some solid tumor cancers, many solid tumors remain resistant to these modes of treatment. In solid tumors, the development of effective antitumor immune responses is hampered by restricted immune cell infiltration and an immunosuppressive tumor microenvironment (TME). An immunotherapy that infiltrates and persists in the solid TME, while providing local, stable levels of therapeutic to activate or reinvigorate antitumor immunity could overcome these challenges faced by current immunotherapies.MethodsUsing lentivirus-driven engineering, we programmed human and murine macrophages to express therapeutic payloads, including Interleukin (IL)-12. In vitro coculture studies were used to evaluate the effect of genetically engineered macrophages (GEMs) secreting IL-12 on T cells and on the GEMs themselves. The effects of IL-12 GEMs on gene expression profiles within the TME and tumor burden were evaluated in syngeneic mouse models of glioblastoma and melanoma and in human tumor slices isolated from patients with advanced gastrointestinal malignancies.ResultsHere, we present a cellular immunotherapy platform using lentivirus-driven genetic engineering of human and mouse macrophages to constitutively express proteins, including secreted cytokines and full-length checkpoint antibodies, as well as cytoplasmic and surface proteins that overcomes these barriers. GEMs traffic to, persist in, and express lentiviral payloads in xenograft mouse models of glioblastoma, and express a non-signaling truncated CD19 surface protein for elimination. IL-12-secreting GEMs activated T cells and induced interferon-gamma (IFNγ) in vitro and slowed tumor growth resulting in extended survival in vivo. In a syngeneic glioblastoma model, IFNγ signaling cascades were also observed in mice treated with mouse bone-marrow-derived GEMs secreting murine IL-12. These findings were reproduced in ex vivo tumor slices comprised of intact MEs. In this setting, IL-12 GEMs induced tumor cell death, chemokines and IFNγ-stimulated genes and proteins.ConclusionsOur data demonstrate that GEMs can precisely deliver titratable doses of therapeutic proteins to the TME to improve safety, tissue penetrance, targeted delivery and pharmacokinetics.
- Published
- 2020
41. Silver linings at the bench and bedside
- Author
-
Venu G. Pillarisetty and Steven C. Katz
- Subjects
Cancer Research ,2019-20 coronavirus outbreak ,COVID-19 Vaccines ,Coronavirus disease 2019 (COVID-19) ,business.industry ,SARS-CoV-2 ,Drug discovery ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Drug development ,Virology ,COVID-19 Drug Treatment ,Correspondence ,Molecular Medicine ,Medicine ,Humans ,business ,Molecular Biology - Published
- 2020
42. Modulation of Immune Checkpoints by Chemotherapy in Human Colorectal Liver Metastases
- Author
-
Leroy Hood, Venu G. Pillarisetty, Qiang Tian, Kevin M. Sullivan, Raymond S. Yeung, Christopher Lausted, Priyanka Baloni, Dani E Bergey, Xiaowei Yan, Changting Meng, Neda Jabbari, and Heidi L. Kenerson
- Subjects
PD-L1 ,Organoplatinum Compounds ,Colorectal cancer ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,immune microenvironment ,colorectal cancer ,Irinotecan ,chemotherapy ,General Biochemistry, Genetics and Molecular Biology ,Article ,galectin-9 ,Immune system ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Tumor Microenvironment ,Humans ,single-cell analysis ,Neoplasm Metastasis ,Hepatitis A Virus Cellular Receptor 2 ,Chemotherapy ,biology ,business.industry ,Liver Neoplasms ,single-cell transcriptome ,medicine.disease ,Immune checkpoint ,digestive system diseases ,Oxaliplatin ,Blockade ,organotypic culture ,Cancer research ,biology.protein ,Camptothecin ,business ,Colorectal Neoplasms ,liver metastases ,TIM3 ,medicine.drug - Abstract
Summary Metastatic colorectal cancer (CRC) is a major cause of cancer-related death, and incidence is rising in younger populations (younger than 50 years). Current chemotherapies can achieve response rates above 50%, but immunotherapies have limited value for patients with microsatellite-stable (MSS) cancers. The present study investigates the impact of chemotherapy on the tumor immune microenvironment. We treat human liver metastases slices with 5-fluorouracil (5-FU) plus either irinotecan or oxaliplatin, then perform single-cell transcriptome analyses. Results from eight cases reveal two cellular subtypes with divergent responses to chemotherapy. Susceptible tumors are characterized by a stemness signature, an activated interferon pathway, and suppression of PD-1 ligands in response to 5-FU+irinotecan. Conversely, immune checkpoint TIM-3 ligands are maintained or upregulated by chemotherapy in CRC with an enterocyte-like signature, and combining chemotherapy with TIM-3 blockade leads to synergistic tumor killing. Our analyses highlight chemomodulation of the immune microenvironment and provide a framework for combined chemo-immunotherapies., Graphical Abstract, Highlights CRLM slice culture can assess immune response to chemotherapy Single-cell analysis identifies cancer subtypes with differing response to chemotherapy 5-FU+irinotecan modulates interferon and PD-L1 pathways in stem-like CRLM Combining chemotherapy with TIM-3 blockade is synergistic in enterocyte-like CRLM, The response of colorectal liver metastases (CRLM) to chemotherapy is analyzed in 3D organotypic tumor slices by single-cell RNA-seq. Jabbari et al. find two subtypes (stem-like and enterocyte-like) of CRLM that express different immune checkpoint ligands and respond differently to chemotherapy. The study highlights the chemomodulation of the tumor immune microenvironment that can be targeted therapeutically.
- Published
- 2020
43. Tumor slice culture as a biologic surrogate of human cancer
- Author
-
Christopher Lausted, Kathryn M. Stadeli, Venu G. Pillarisetty, Kevin M. Sullivan, Matthew M. Yeh, Raymond S. Yeung, Heidi L. Kenerson, James O. Park, Kimberly J. Riehle, Qiang Tian, Yongwoo D. Seo, Xiaowei Yan, and Cigdem Himmetoglu Ussakli
- Subjects
0301 basic medicine ,Tumor microenvironment ,Pathology ,medicine.medical_specialty ,business.industry ,Cea mrna ,General Medicine ,Liver resections ,In vitro ,03 medical and health sciences ,Vibratome ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Medicine ,Tumor necrosis factor alpha ,Original Article ,business ,Ex vivo ,Human cancer - Abstract
Background The tumor microenvironment (TME) is critical to every aspect of cancer biology. Organotypic tumor slice cultures (TSCs) preserve the original TME and have demonstrated utility in predicting drug sensitivity, but the association between clinicopathologic parameters and in vitro TSC behavior has not been well-defined. Methods One hundred and eight fresh tumor specimens from liver resections at a tertiary academic center were procured and precisely cut with a Vibratome to create 250 μm × 6 mm slices. These fixed-dimension TSCs were grown on polytetrafluoroethylene inserts, and their metabolic activities were determined by a colorimetric assay. Correlation between baseline activities and clinicopathologic parameters was assessed. Tissue CEA mRNA expression was determined by RNAseq. Results By standardizing the dimensions of a slice, we found that adjacent tumor slices have equivalent metabolic activities, while those derived from different tumors exhibit >30-fold range in baseline MTS absorbances, which correlated significantly with the percentage of tumor necrosis based on histologic assessment. Extending this to individual cancers, we were able to detect intra-tumoral heterogeneity over a span of a few millimeters, which reflects differences in tumor cell density and Ki-67 positivity. For colorectal cancers, tissue CEA expression based on RNAseq of tumor slices was found to correlate with clinical response to chemotherapies. Conclusions We report a standardized method to assess and compare human cancer growth ex vivo across a wide spectrum of tumor samples. TSC reflects the state of tumor behavior and heterogeneity, thus providing a simple approach to study of human cancers with an intact TME.
- Published
- 2020
44. 661 Neoantigen-specific CD4+ T cells in human melanoma have diverse differentiation states and correlate with CD8+ T cell, macrophage, and B cell function
- Author
-
Evan W. Newell, Venu G. Pillarisetty, David R. Byrd, Shailender Bhatia, Ata S. Moshiri, Raphael Gottardo, Julia Szeto, Naina Singhi, Joshua R. Veatch, Scott S. Tykodi, Kimberly S. Smythe, John F. Thompson, Evan T. Hall, Sylvia Lee, Carolyn Shasha, Stanley R. Riddell, and Teresa S. Kim
- Subjects
Pharmacology ,Cancer Research ,Chemistry ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.anatomical_structure ,Oncology ,medicine ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Macrophage ,Cytotoxic T cell ,Human melanoma ,RC254-282 ,B cell ,Function (biology) - Abstract
BackgroundTumor-antigen specific CD4+ T cells are crucial for the efficacy of antibodies that block immune checkpoint proteins in mouse tumor models, but their activities in human tumor immunity are less clear. CD8+ T cells infiltrating human tumors, including those specific for tumor antigens, have been studied using single cell profiling techniques and exist in a variety of dysfunctional states. The transcriptional states of tumor-specific CD4+ T cells present in tumors and their potential contributions to the tumor microenvironment are less well understood.MethodsWe used targeted single cell RNA sequencing and matching of T cell receptor (TCR) sequences to identify phenotypic signatures that discriminated tumor antigen- and viral antigen-specific CD4+ T cells infiltrating human melanoma tumors in four patients. The presence of CD4+ T cells with these signatures was correlated with the number and phenotype of other immune cells in the tumor microenvironment in an extended cohort of 20 patients.ResultsWe identified 259 CD4+ T cells representing 40 different TCR clonotypes specific for 13 neoantigens and 108 cells representing 14 TCR clonotypes specific for self-antigens in four melanoma patients. High expression of CXCL13 defined conventional CD4+ T cells that recognize tumor associated neoantigens and self-antigens from bystander and viral antigen-specific CD4+ T cells. Tumor-reactive CD4+ T cells could be subdivided into clusters expressing memory and T follicular helper markers, and those expressing cytolytic markers and IFN-g. In an extended cohort of 20 patients with melanoma, the frequency of CXCL13+ CD4+ T cells in the tumor microenvironment correlated with the presence and proliferation of CD8+ T cells, the presence and maturation of B cells, the activation of interferon responsive genes in tumor associated macrophages, and patient survival. CD4+ T cells with similar transcriptional signatures were identified in data sets from breast and non-small cell lung cancer, suggesting these markers may enrich for tumor-reactive CD4+ T cells in many cancers.ConclusionsThese results identify a subset of tumor infiltrating conventional CD4+ T cells in melanoma that are enriched for reactivity to tumor antigens and exist in multiple phenotypic states. Correlations of the presence of these cells with the frequency and phenotype of other immune cells suggest roles for these tumor antigen-specific CD4+ T cells in providing CD8+ T cell help, driving recruitment and maturation of B cells, and activating macrophages. Isolating such cells based on their unique phenotype and utilizing them for adoptive therapy could alter the tumor microenvironment for therapeutic benefit.Ethics ApprovalAll Patient samples in this study were obtained from patients who signed informed consent in a study approved by the institutional review board of the Fred Hutchinson Cancer Research Center (protocol #2643).
- Published
- 2021
45. Lanreotide for the prevention of postoperative pancreatic fistula: phase II clinical trial results
- Author
-
J. Park, Venu G. Pillarisetty, A. Abbasi, and J. Sham
- Subjects
Clinical trial ,medicine.medical_specialty ,chemistry.chemical_compound ,Hepatology ,chemistry ,business.industry ,Pancreatic fistula ,Gastroenterology ,Medicine ,business ,Lanreotide ,medicine.disease ,Surgery - Published
- 2021
46. Abstract 607: IL-15 is the most potent of tested gamma chain cytokines at inducing in situ proliferation of T cells in human pancreatic cancer
- Author
-
Venu G. Pillarisetty, Cynthia Hsu, Xiuyun Jiang, Shihong Zhang, Arezou Abbasi, Teresa S. Kim, and Karan Kohli
- Subjects
In situ ,Cancer Research ,Oncology ,Chemistry ,Interleukin 15 ,Pancreatic cancer ,medicine ,Cancer research ,medicine.disease - Abstract
Background: Multiple factors, including dense stroma and high infiltration of suppressive immune cells make pancreatic ductal adenocarcinoma (PDA) challenging to treat with immunotherapy. The presence of intra-tumoral T cells positively correlates with improved survival for PDA patients. Gamma chain cytokines (GCCs) can augment T cell mediated anti-tumor immunity and various GCC agonist drugs have been applied in clinical trials for cancer. Besides evaluating the effect of GCC derived agonists using recovered blood or biopsies of treated patients, it is challenging to mechanistically study the effect of GCCs on tumor-resident immune cells in human samples. Methods: Here we apply organotypic slice culture of surgically resected tumor tissues to study the effect of GCCs on intra-tumoral immune cells. We treated slices of PDAs with GCCs (IL-2, IL-7, IL-15 and IL-21) for 6 days. Immune cells, especially T cells emigrated from the tumor slices into the culture supernatant. On day 6 we fixed intact tumor slices for multiplex immunohistochemistry (mIHC) and analyzed emigrated cells using flow cytometry. Results: IL-15 and IL-7 potently induced T cell proliferation. The effect of IL-7 and IL-15 on T cell proliferation was higher for T cells that expressed markers of antigen experience such as CD39 and PD1 (Table 1). While IL-15 had the strongest effect on tumor-derived T cells, IL-15 and IL-7 were similar in their effect on T cells in the blood of the same patient, suggesting that IL-15 is especially effective in inducing the proliferation of tumor-infiltrating T cells. The effect of IL-15 on enhanced T cell proliferation was also seen within tumor slices, as revealed by mIHC. Conclusion: Our data suggest that IL-15 is the most potent GCC at inducing in situ expansion of tumor-resident T cells, including T cells that show signs of antigen experience. Table 1.IL-15 induces in situ T cell proliferation in PDA.AssayGroupsUntreatedIL-2IL-7IL-15IL-21ParameterFlow Cytometry%Ki67+CD8+cells of live cells0.090.040.360.595.001.3514.331.200.310.14%CD39+CD103+of CD8+0.130.150.110.040.290.140.620.180.130.08%PD1+of CD8+25.107.2223.233.1030.484.7446.483.5037.402.32mIHC%CD8+Ki67+ of all cells0.16 0.04(n=2)0.83 (n=1)0.550.38(n=2)2.200.04(n=2)0.490.41(n=2)n=4 slices for each group for flow cytometry. Data are from one experiment*. MeanSD is shown. Two doses (10-fold difference) for each cytokine were tested and results only for the low dose are shown.*Another experiment with groups IL-2 and IL-2+IL-15 was performed and had similar results for the effect of IL-15 Citation Format: Karan Kohli, Shihong Zhang, Xiuyun Jiang, Cynthia Hsu, Arezou Abbasi, Teresa S. Kim, Venu G. Pillarisetty. IL-15 is the most potent of tested gamma chain cytokines at inducing in situ proliferation of T cells in human pancreatic cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 607.
- Published
- 2021
47. Focused ultrasound for immuno-adjuvant treatment of pancreatic cancer: An emerging clinical paradigm in the era of personalized oncotherapy
- Author
-
Ezekiel Maloney, Venu G. Pillarisetty, T.D. Khokhlova, Lorenzo Giuliani, Matteo Primavera, Yak-Nam Wang, Joo Ha Hwang, George R. Schade, and Elizabeth A. Repasky
- Subjects
Cytotoxicity, Immunologic ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Ultrasonic Therapy ,medicine.medical_treatment ,T cell ,Immunology ,Lymphocyte Activation ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Adjuvants, Immunologic ,Pancreatic cancer ,medicine ,Animals ,Humans ,Immunology and Allergy ,Precision Medicine ,business.industry ,Cancer ,Cell Differentiation ,Immunotherapy ,Dendritic cell ,medicine.disease ,Precision medicine ,Carcinoma, Ductal ,Pancreatic Neoplasms ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Immunization ,business ,Adjuvant - Abstract
Current clinical treatment regimens, including many emergent immune strategies (e.g., checkpoint inhibitors) have done little to affect the devastating course of pancreatic ductal adenocarcinoma (PDA). Clinical trials for PDA often employ multi-modal treatment, and have started to incorporate stromal-targeted therapies, which have shown promising results in early reports. Focused ultrasound (FUS) is one such therapy that is uniquely equipped to address local and systemic limitations of conventional cancer therapies as well as emergent immune therapies for PDA. FUS methods can non-invasively generate mechanical and/or thermal effects that capitalize on the unique oncogenomic/proteomic signature of a tumor. Potential benefits of FUS therapy for PDA include: (1) emulsification of targeted tumor into undenatured antigens in situ, increasing dendritic cell maturation, and increasing intra-tumoral CD8+/ T regulatory cell ratio and CD8+ T cell activity; (2) reduction in intra-tumoral hypoxic stress; (3) modulation of tumor cell membrane protein localization to enhance immunogenicity; (4) modulation of the local cytokine milieu toward a Th1-type inflammatory profile; (5) up-regulation of local chemoattractants; (6) remodeling the tumor stroma; (7) localized delivery of exogenously packaged immune-stimulating antigens, genes and therapeutic drugs. While not all of these results have been studied in experimental PDA models to date, the principles garnered from other solid tumor and disease models have direct relevance to the design of optimal FUS protocols for PDA. In this review, we address the pertinent limitations in current and emergent immune therapies that can be improved with FUS therapy for PDA.
- Published
- 2017
48. Disrupting glutamine metabolic pathways to sensitize gemcitabine-resistant pancreatic cancer
- Author
-
Lisa A. Lai, Lei Wang, Jonah Riddell, Venu G. Pillarisetty, Linda Jung, Ru Chen, Teresa A. Brentnall, Yumi Sullivan, Melissa Wong, and Sheng Pan
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Proteomics ,Cell Survival ,Glutamine ,Science ,Diazooxonorleucine ,Biology ,Deoxycytidine ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Biosynthesis ,Pancreatic cancer ,Cell Line, Tumor ,medicine ,Humans ,Cell Proliferation ,Multidisciplinary ,Cell growth ,Drug Synergism ,medicine.disease ,Gemcitabine ,3. Good health ,Pancreatic Neoplasms ,Metabolic pathway ,030104 developmental biology ,Biochemistry ,chemistry ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Medicine ,Oxidation-Reduction ,Metabolic Networks and Pathways ,medicine.drug - Abstract
Pancreatic cancer is a lethal disease with poor prognosis. Gemcitabine has been the first line systemic treatment for pancreatic cancer. However, the rapid development of drug resistance has been a major hurdle in gemcitabine therapy leading to unsatisfactory patient outcomes. With the recent renewed understanding of glutamine metabolism involvement in drug resistance and immuno-response, we investigated the anti-tumor effect of a glutamine analog (6-diazo-5-oxo-L-norleucine) as an adjuvant treatment to sensitize chemoresistant pancreatic cancer cells. We demonstrate that disruption of glutamine metabolic pathways improves the efficacy of gemcitabine treatment. Such a disruption induces a cascade of events which impacts glycan biosynthesis through Hexosamine Biosynthesis Pathway (HBP), as well as cellular redox homeostasis, resulting in global changes in protein glycosylation, expression and functional effects. The proteome alterations induced in the resistant cancer cells and the secreted exosomes are intricately associated with the reduction in cell proliferation and the enhancement of cancer cell chemosensitivity. Proteins associated with EGFR signaling, including downstream AKT-mTOR pathways, MAPK pathway, as well as redox enzymes were downregulated in response to disruption of glutamine metabolic pathways.
- Published
- 2017
49. T-cell infiltration and clonality correlate with programmed cell death protein 1 and programmed death-ligand 1 expression in patients with soft tissue sarcomas
- Author
-
Wendy M. Blumenschein, Kelsey Baker, Bailey Donahue, Jennifer H. Yearley, Terrill K. McClanahan, Benjamin Hoch, Erin Murphy, Yuzheng Zhang, Marissa Vignali, Lee D. Cranmer, Stanley R. Riddell, Robin L. Jones, Sara Cooper, Matthew B. Spraker, Ryan O. Emerson, Seth M. Pollack, Steven M. Townson, Elizabeth T. Loggers, Qianchuan He, Y. David Seo, Venu G. Pillarisetty, Sharon Benzeno, Mary W. Redman, and Robert W. Ricciotti
- Subjects
0301 basic medicine ,Leiomyosarcoma ,Cancer Research ,Pathology ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Soft tissue sarcoma ,Immunotherapy ,Round Cell Liposarcoma ,Liposarcoma ,medicine.disease ,Undifferentiated Pleomorphic Sarcoma ,Synovial sarcoma ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Sarcoma ,business - Abstract
BACKGROUND Patients with metastatic sarcomas have poor outcomes and although the disease may be amenable to immunotherapies, information regarding the immunologic profiles of soft tissue sarcoma (STS) subtypes is limited. METHODS The authors identified patients with the common STS subtypes: leiomyosarcoma, undifferentiated pleomorphic sarcoma (UPS), synovial sarcoma (SS), well-differentiated/dedifferentiated liposarcoma, and myxoid/round cell liposarcoma. Gene expression, immunohistochemistry for programmed cell death protein (PD-1) and programmed death-ligand 1 (PD-L1), and T-cell receptor Vβ gene sequencing were performed on formalin-fixed, paraffin-embedded tumors from 81 patients. Differences in liposarcoma subsets also were evaluated. RESULTS UPS and leiomyosarcoma had high expression levels of genes related to antigen presentation and T-cell infiltration. UPS were found to have higher levels of PD-L1 (P≤.001) and PD-1 (P≤.05) on immunohistochemistry and had the highest T-cell infiltration based on T-cell receptor sequencing, significantly more than SS, which had the lowest (P≤.05). T-cell infiltrates in UPS also were more oligoclonal compared with SS and liposarcoma (P≤.05). A model adjusted for STS histologic subtype found that for all sarcomas, T-cell infiltration and clonality were highly correlated with PD-1 and PD-L1 expression levels (P≤.01). CONCLUSIONS In the current study, the authors provide the most detailed overview of the immune microenvironment in sarcoma subtypes to date. UPS, which is a more highly mutated STS subtype, provokes a substantial immune response, suggesting that it may be well suited to treatment with immune checkpoint inhibitors. The SS and liposarcoma subsets are less mutated but do express immunogenic self-antigens, and therefore strategies to improve antigen presentation and T-cell infiltration may allow for successful immunotherapy in patients with these diagnoses. Cancer 2017;123:3291-304. © 2017 The Authors. Cancer published by Wiley Periodicals, Inc. on behalf of American Cancer Society. This is an open access article under the terms of the Creative Commons Attribution NonCommercial License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes.
- Published
- 2017
50. Biopolymers codelivering engineered T cells and STING agonists can eliminate heterogeneous tumors
- Author
-
Smitha P. S. Pillai, Sirkka B. Stephan, Venu G. Pillarisetty, Xiuyun Jiang, Tyrel T. Smith, K. Dane Wittrup, Cary F. Opel, Howell F. Moffett, Amy Dumigan, and Matthias Stephan
- Subjects
0301 basic medicine ,Adoptive cell transfer ,T-Lymphocytes ,T cell ,Melanoma, Experimental ,Antigen-Presenting Cells ,Antineoplastic Agents ,Mice, Transgenic ,Biology ,03 medical and health sciences ,Biopolymers ,0302 clinical medicine ,Immune system ,Implants, Experimental ,Cell Line, Tumor ,Pancreatic cancer ,medicine ,Animals ,Receptor ,Cyclic GMP ,Drug Carriers ,Melanoma ,Membrane Proteins ,General Medicine ,medicine.disease ,Adoptive Transfer ,Chimeric antigen receptor ,3. Good health ,Mice, Inbred C57BL ,Pancreatic Neoplasms ,030104 developmental biology ,medicine.anatomical_structure ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Neoplasm Transplantation ,Carcinoma, Pancreatic Ductal ,Research Article - Abstract
Therapies using T cells that are programmed to express chimeric antigen receptors (CAR T cells) consistently produce positive results in patients with hematologic malignancies. However, CAR T cell treatments are less effective in solid tumors for several reasons. First, lymphocytes do not efficiently target CAR T cells; second, solid tumors create an immunosuppressive microenvironment that inactivates T cell responses; and third, solid cancers are typified by phenotypic diversity and thus include cells that do not express proteins targeted by the engineered receptors, enabling the formation of escape variants that elude CAR T cell targeting. Here, we have tested implantable biopolymer devices that deliver CAR T cells directly to the surfaces of solid tumors, thereby exposing them to high concentrations of immune cells for a substantial time period. In immunocompetent orthotopic mouse models of pancreatic cancer and melanoma, we found that CAR T cells can migrate from biopolymer scaffolds and eradicate tumors more effectively than does systemic delivery of the same cells. We have also demonstrated that codelivery of stimulator of IFN genes (STING) agonists stimulates immune responses to eliminate tumor cells that are not recognized by the adoptively transferred lymphocytes. Thus, these devices may improve the effectiveness of CAR T cell therapy in solid tumors and help protect against the emergence of escape variants.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.