Journal of Immunotherapy for Cancer最新文献

筛选
英文 中文
Correction: Early disappearance of tumor antigen-reactive T cells from peripheral blood correlates with superior clinical outcomes in melanoma under anti-PD-1 therapy 更正:外周血中肿瘤抗原反应性T细胞的早期消失与抗pd -1治疗下黑色素瘤的良好临床结果相关
Journal of Immunotherapy for Cancer Pub Date : 2022-04-01 DOI: 10.1136/jitc-2021-003439corr1
{"title":"Correction: Early disappearance of tumor antigen-reactive T cells from peripheral blood correlates with superior clinical outcomes in melanoma under anti-PD-1 therapy","authors":"","doi":"10.1136/jitc-2021-003439corr1","DOIUrl":"https://doi.org/10.1136/jitc-2021-003439corr1","url":null,"abstract":"","PeriodicalId":16067,"journal":{"name":"Journal of Immunotherapy for Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"85846976","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
P855 High-resolution maps of heterogeneous antigen expression in glioblastoma and implications for immunotherapy 胶质母细胞瘤中异质抗原表达的高分辨率图谱及其免疫治疗的意义
Journal of Immunotherapy for Cancer Pub Date : 2020-04-01 DOI: 10.1136/LBA2019.10
M. Barish, L. Weng, Dina Awabdeh, Blake Brewster, M. D’Apuzzo, Yubo Zhai, Alfonso Brito, B. Chang, Annie Sarkissian, R. Starr, W. Chang, B. Aguilar, A. Naranjo, S. Blanchard, Russell C. Rockne, B. Badie, Vanessa Jonsson, Stephen J. Forman, Christine Brown
{"title":"P855 High-resolution maps of heterogeneous antigen expression in glioblastoma and implications for immunotherapy","authors":"M. Barish, L. Weng, Dina Awabdeh, Blake Brewster, M. D’Apuzzo, Yubo Zhai, Alfonso Brito, B. Chang, Annie Sarkissian, R. Starr, W. Chang, B. Aguilar, A. Naranjo, S. Blanchard, Russell C. Rockne, B. Badie, Vanessa Jonsson, Stephen J. Forman, Christine Brown","doi":"10.1136/LBA2019.10","DOIUrl":"https://doi.org/10.1136/LBA2019.10","url":null,"abstract":"Background Glioblastoma (GBM) remains an almost universally fatal brain tumor. While CAR T cell immunotherapy has shown promising clinical efficacy, therapeutic failure may reflect our incomplete understanding of target antigen expression. We previously examined variations in antigen expression at the level of individual patients (inter-patient or inter-tumor heterogeneity), focusing on immunotherapy targets IL13Rα2, HER2 and EGFR. We concluded that antigen expression diverged from expectations from random expression. Because antigen escape may arise from GBM cell heterogeneity, we have mapped target antigen expression within individual tumors (intra-tumor heterogeneity). Methods Serial sections from a 43 patient cohort were immunostained (DAB with hematoxylin counterstain) for target antigens IL13Rα2, HER2 and EGFR. Each section was annotated directly from the slide by a neuropathologist. Sections were scanned (0.46 µm/pixel; Hamamatsu), and then working within Fiji/ImageJ, images were segmented by color deconvolution into hematoxylin (nuclei) and DAB layers. Images of nuclear layers were aligned, and used to align the DAB layers. Two schemes were used to examine the spatial distributions of the three target antigens. When tumor domains could be identified, we determined expression of each antigen as optical density (OD). In the second scheme, a 10 µm grid was superimposed on each section, and OD was determined for each position and assembled into spreadsheets (Origin v2019b). Maps for expression were generated from the OD in each position. Results Approaching these maps from the perspective of antigen escape, we examined the extent to which expression of target antigens was spatially mixed, how rapidly antigen dominance could shift (spatial frequency), and whether spatial distributions were arrayed in a coordinated manner. When tumor domains could be identified, we calculated the Shannon diversity index (H) for each domain within a section. While values of H clustered within some tumors, usually values of H varied widely. The superimposed grid was used to examine heterogeneity within entire tumor sections. Expression was intermixed, and EGFR and IL13Rα2/HER2 displayeds complementary expression patterns. In tumors with large EGFR+ areas, IL13Rα2+/HER2+ areas could overlap, while when EGFR+ areas were smaller, IL13Rα2+ and HER2+ areas were more distinct. Borders could be quite diffuse, or quite sharp (a few cell diameters). Conclusions Our results indicate that expression of IL13Rα2, HER2 and EGFR is highly heterogeneous and not always spatially distinct. Because GBM tumors adapt to the selection pressures of immunotherapies, we suggest that combination therapies should be designed accordingly, and immunotherapies targeting IL13Rα2/HER2 could benefit from inclusion of EGFR.","PeriodicalId":16067,"journal":{"name":"Journal of Immunotherapy for Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"81504387","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 7
P860 Results from a combination of OX40 (PF-04518600) and 4–1BB (utomilumab) agonistic antibodies in melanoma and non-small cell lung cancer in a phase 1 dose expansion cohort 在1期剂量扩大队列中,OX40 (PF-04518600)和4-1BB (utomilumab)激动抗体联合治疗黑色素瘤和非小细胞肺癌的结果
Journal of Immunotherapy for Cancer Pub Date : 2020-04-01 DOI: 10.1136/LBA2019.14
A. Chiappori, John A Thompson, F. Eskens, J. Spano, T. Doi, O. Hamid, A. Diab, N. Rizvi, S. Hu-Lieskovan, W. Ros, Jacob S. Thomas, A. Forgie, Wenjing Yang, K. Liao, Ray Li, Farhad Kazazi, J. Chou, A. E. Khoueiry
{"title":"P860 Results from a combination of OX40 (PF-04518600) and 4–1BB (utomilumab) agonistic antibodies in melanoma and non-small cell lung cancer in a phase 1 dose expansion cohort","authors":"A. Chiappori, John A Thompson, F. Eskens, J. Spano, T. Doi, O. Hamid, A. Diab, N. Rizvi, S. Hu-Lieskovan, W. Ros, Jacob S. Thomas, A. Forgie, Wenjing Yang, K. Liao, Ray Li, Farhad Kazazi, J. Chou, A. E. Khoueiry","doi":"10.1136/LBA2019.14","DOIUrl":"https://doi.org/10.1136/LBA2019.14","url":null,"abstract":"Background PF-04518600 (PF-8600) and utomilumab (uto) are humanized agonist IgG2 monoclonal antibodies for the tumor necrosis factor superfamily receptors OX40 and 4-1BB, respectively. In a phase I dose escalation study (NCT02315066), this antibody combination was tolerable at all dose levels and induced responses in patients with melanoma resistant to immune checkpoint inhibitors. We report results from a dose expansion cohort of this study of patients with melanoma and non-small cell lung cancer (NSCLC) treated with PF-8600 (OX40 antibody) in combination with uto. Efficacy, safety, and the association of baseline and pharmacodynamic biomarkers with efficacy were examined. Methods In this expansion cohort, patients with locally advanced/metastatic melanoma (n=10) and NSCLC (n=20) who progressed on prior anti-PD1/PD-L1 treatment and/or anti CTLA4 treatment (melanoma only) were enrolled. Patients received OX40 antibody 30 mg IV every 2wks in combination with uto 20 mg IV every 28d. Tumor assessments were performed every 8wks using RECIST1.1. Paired biopsy samples collected at baseline and 6wks were analyzed by immunohistochemistry and RNA sequencing to evaluate the pharmacodynamic effects of the OX40 antibody in combination with uto. Whole blood samples were collected longitudinally, from which DNA was extracted and submitted for high-throughput sequencing of the T cell receptor β-chain. Results One patient with NSCLC achieved a confirmed and ongoing partial response lasting at least 6 months; Based on analyses of a subset of baseline biopsies, this patient’s tumor had the lowest FOXP3 expression. A total of 7 (70%) melanoma patients and 7 (35%) NSCLC patients achieved a best overall response of stable disease (SD). The median duration of SD was 16.3 weeks (melanoma: 16.0 weeks; NSCLC: 24.1 weeks), for a disease control rate of 50%. Among patients with a defined response, paired biopsy analyses showed that the greatest increase in CD8 occurred in the NSCLC patient with the longest duration of stable disease. The most frequent treatment related adverse events (TRAEs) reported in ≥10% of patients were pruritis, anemia, fatigue, decreased appetite, and rash. Grade 3 TRAEs, rash and lymphocyte count decreased, were reported in 5 patients and a grade 4 TRAE of lipase increased (asymptomatic) was reported in 1 patient. Conclusions The combination of PF-8600 and uto had a tolerable safety profile and demonstrated clinical benefit, including in an NSCLC patient who had progressed on anti-PD1 therapy and achieved a durable partial response. Further combinations with one or both of these immune costimulatory receptor agonist antibodies might enhance their efficacy. Acknowledgements This study was funded by Pfizer Inc. Editorial support was provided by Chu Kong Liew, PhD, of Engage Scientific Solutions and was funded by Pfizer Inc. Trial Registration ClinicalTrials. gov: NCT02315066 Ethics Approval The study was approved by the institutional review board at ","PeriodicalId":16067,"journal":{"name":"Journal of Immunotherapy for Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"81529013","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 9
O81 IMpower110: interim overall survival (OS) analysis of a phase III study of atezolizumab (ATEZO) monotherapy vs platinum-based chemotherapy (CHEMO) as first-line (1L) treatment in PD-L1–selected NSCLC O81 IMpower110:一项III期研究的中期总生存期(OS)分析:atezolizumab (ATEZO)单药治疗与铂基化疗(CHEMO)作为pd - l1选择的非小细胞肺癌一线(1L)治疗
Journal of Immunotherapy for Cancer Pub Date : 2020-04-01 DOI: 10.1136/LBA2019.1
R. Herbst, F. Marinis, G. Giaccone, N. Reinmuth, A. Vergnenègre, C. Barrios, M. Morise, E. Font, Z. Andrić, Sarayut Lucien Geater, M. Ozguroglu, S. Mocci, M. McCleland, I. Enquist, K. Komatsubara, Y. Deng, H. Kuriki, X. Wen, J. Jassem, D. Spigel
{"title":"O81 IMpower110: interim overall survival (OS) analysis of a phase III study of atezolizumab (ATEZO) monotherapy vs platinum-based chemotherapy (CHEMO) as first-line (1L) treatment in PD-L1–selected NSCLC","authors":"R. Herbst, F. Marinis, G. Giaccone, N. Reinmuth, A. Vergnenègre, C. Barrios, M. Morise, E. Font, Z. Andrić, Sarayut Lucien Geater, M. Ozguroglu, S. Mocci, M. McCleland, I. Enquist, K. Komatsubara, Y. Deng, H. Kuriki, X. Wen, J. Jassem, D. Spigel","doi":"10.1136/LBA2019.1","DOIUrl":"https://doi.org/10.1136/LBA2019.1","url":null,"abstract":"Background PD-L1/PD-1 inhibitors (CPI) as monotherapy or in combination with platinum-based doublet chemo (± bevacizumab) are 1L treatment options in metastatic NSCLC, with choice of agent(s) determined by PD-L1 expression. For patients (pts) who may be ineligible for combination therapy, CPI monotherapy remains an attractive treatment choice. IMpower110 evaluated atezo as 1L treatment in PD-L1–selected pts independent of tumor histology. Methods IMpower110 enrolled 572 chemo-naive pts with stage IV nonsquamous (nsq) or squamous (sq) NSCLC, PD-L1 expression ≥ 1% on TC or IC, measurable disease by RECIST 1.1 and ECOG PS 0-1. Pts were randomized 1:1 to receive atezo 1200 mg IV q3w (Arm A) or platinum-based chemo (Arm B; 4 or 6 21-day cycles). Arm B nsq pts received cisplatin (cis) 75 mg/m2 or carboplatin (carbo) AUC 6 + pemetrexed 500 mg/m2 IV q3w; Arm B sq pts received cis 75 mg/m2 + gemcitabine (gem) 1250 mg/m2 or carbo AUC 5 + gem 1000 mg/m2 IV q3w. Stratification factors were sex, ECOG PS, histology and tumor PD-L1 status (TC1/2/3 and any IC vs TC0 and IC1/2/3). The primary endpoint of OS is tested hierarchically in the wild-type (WT; EGFR/ALK-negative) population (TC3 or IC3 then TC2/3 or IC2/3 then TC1/2/3 or IC1/2/3). Results The 3 primary efficacy populations included 554 TC1/2/3 or IC1/2/3 WT pts, 328 TC2/3 or IC2/3 WT pts and 205 TC3 or IC3 WT pts. Median follow-up was 15.7 months (range, 0-35) in TC3 or IC3 WT pts. In the TC3 or IC3 WT population, atezo monotherapy improved median OS by 7.1 months (HR, 0.595; P = 0.0106) compared with chemo (table 1). The safety population comprised 286 pts in Arm A and 263 in Arm B. Treatment-related AEs (TRAEs) and Grade 3-4 TRAEs occurred in 60.5% (Arm A) and 85.2% (Arm B), and 12.9% (Arm A) and 44.1% (Arm B), respectively. Abstract 081 Table 1 Conclusions At this interim analysis, IMpower110 met the primary endpoint of OS with statistically significant and clinically meaningful improvement in the TC3 or IC3 WT population. The safety profile favored Arm A, with no new or unexpected safety signals identified. Trial Registration NCT02409342 Ethics Approval The trial was conducted according to the principles of the Declaration of Helsinki. All patients provided written informed consent. Protocol approval was obtained from independent review boards or ethics committees at each site.","PeriodicalId":16067,"journal":{"name":"Journal of Immunotherapy for Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"84847436","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 7
O85 Durable responses in anti-PD-1 refractory melanoma following intratumoral injection of a toll-like receptor 9 (TLR9) agonist, CMP-001, in combination with pembrolizumab 肿瘤内注射toll样受体9 (TLR9)激动剂CMP-001联合派姆单抗治疗抗pd -1难治性黑色素瘤的持久疗效
Journal of Immunotherapy for Cancer Pub Date : 2020-04-01 DOI: 10.1136/LBA2019.4
M. Milhem, Y. Zakharia, D. Davar, E. Buchbinder, T. Medina, A. Daud, A. Ribas, J. Niu, G. Gibney, K. Margolin, A. Olszanski, Interjit Mehmi, Takami Sato, M. Shaheen, Aaron H. Morris, D. Mauro, Katie M. Campbell, R. Bao, G. Weiner, J. Luke, A. Krieg, J. Kirkwood
{"title":"O85 Durable responses in anti-PD-1 refractory melanoma following intratumoral injection of a toll-like receptor 9 (TLR9) agonist, CMP-001, in combination with pembrolizumab","authors":"M. Milhem, Y. Zakharia, D. Davar, E. Buchbinder, T. Medina, A. Daud, A. Ribas, J. Niu, G. Gibney, K. Margolin, A. Olszanski, Interjit Mehmi, Takami Sato, M. Shaheen, Aaron H. Morris, D. Mauro, Katie M. Campbell, R. Bao, G. Weiner, J. Luke, A. Krieg, J. Kirkwood","doi":"10.1136/LBA2019.4","DOIUrl":"https://doi.org/10.1136/LBA2019.4","url":null,"abstract":"Background Intratumoral (IT) injection of CMP-001, a CpG-A TLR9 agonist packaged within a virus-like particle, is designed to activate tumor-associated plasmacytoid dendritic cells, inducing an interferon-rich tumor microenvironment and anti-tumor CD8+ T cell responses. Methods CMP-001-001 is an ongoing Phase 1b trial evaluating the safety and efficacy of CMP-001 in combination with pembrolizumab (Part 1; N = 144) or alone (Part 2; N = 23) in patients with advanced melanoma resistant to prior anti-PD-1 therapy (Tables 1). CMP-001 is administered IT into accessible lesion(s) either with, or without on-site saline dilution (table 1), and response assessed by RECIST v1.1. Monotherapy patients who progress can be rolled over onto combination therapy and continue on study. Baseline and on-therapy serum is analyzed for cytokines, and immunohistochemistry and RNA-Seq are performed on available tumor biopsies. Abstract O85 Table 1 Advanced anti-PD-1 Refractory melanoma patient population by treatment allocation Results Adverse events (AEs) attributed to CMP-001 in combination with pembrolizumab or as monotherapy consisted predominately of transient low-Grade flu-like symptoms and injection site reactions: Grade 3+ related AEs were reported in 33% of patients treated with combination therapy and 22% of patients with monotherapy. The Objective Response Rate (ORR) with undiluted CMP-001 in combination with pembrolizumab was 24% (18/75; 95% confidence interval: 15%-35% (table 1); on-site dilution of CMP-001 was associated with a substantial decrease in ORR to 12% (7/61; 95% confidence interval: 5%-22% (table 1). Three additional patients had a delayed partial response after an initial period of disease progression. Anti-tumor response was comparable between injected and uninjected lesions. The median duration of response to combination therapy has not been reached. The ORR to CMP-001 monotherapy was 22% (5/23; 95% confidence interval: 7%-44% (table 1); time from last anti-PD-1 therapy before CMP-001 was 1.5 to >20 months in responders; 3 of the patients responding to CMP-001 monotherapy achieved PR at the first evaluation, but progressed by the second evaluation. Serum and tumor biopsy translational studies in the patients receiving combination therapy supported the proposed mechanism of TLR9 activation and identified a possible association between induction of serum CXCL10 and response. Conclusions IT CMP-001 alone and in combination with pembrolizumab appears well tolerated, can reverse resistance to anti-PD-1 therapy, and can produce deep and durable clinical responses in patients with advanced melanoma. Ethics Approval CMP-001-001 was centrally approved by the WCG-WIRB, WIRB approval tracking number 20152597.","PeriodicalId":16067,"journal":{"name":"Journal of Immunotherapy for Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"85616365","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 18
P857 ONM-500 – a novel STING-activating therapeutic nanovaccine platform for cancer immunotherapy P857 ONM-500 -一种新的sting激活治疗性纳米疫苗平台,用于癌症免疫治疗
Journal of Immunotherapy for Cancer Pub Date : 2020-04-01 DOI: 10.1136/LBA2019.11
Jason B Miller, Min Luo, Hua Wang, Zhaohui Wang, Xinliang Ding, Ashley Campbell, Jonathan Almazan, Zhijian J. Chen, Jinming Gao, T. Zhao
{"title":"P857 ONM-500 – a novel STING-activating therapeutic nanovaccine platform for cancer immunotherapy","authors":"Jason B Miller, Min Luo, Hua Wang, Zhaohui Wang, Xinliang Ding, Ashley Campbell, Jonathan Almazan, Zhijian J. Chen, Jinming Gao, T. Zhao","doi":"10.1136/LBA2019.11","DOIUrl":"https://doi.org/10.1136/LBA2019.11","url":null,"abstract":"Background Efficacy of cancer vaccines requires the induction of tumor antigen-specific cytotoxic T-lymphocytes (CTL) to effectively clear established tumors. Orchestration of antigen presentation, co-stimulatory signaling, and innate cytokine signals are necessary steps for tumor-specific T-cell activation. The ONM-500 nanovaccine platform1-2 utilizes a novel pH-sensitive polymer that forms an antigen-encapsulating nanoparticle and functions both as a carrier for antigen delivery of both peptide and protein antigens to dendritic cells and acts as an adjuvant, activating the stimulator on interferon genes (STING) pathway and generating a CD8+ CTL response. Peptide antigens have translational challenges due to complex formulations and/or HLA-type-specific antigen sequence recognition, processing and presentation. Full-length protein antigens alleviate HLA subtype limitation, allowing coverage of multi-immunogenic T cell epitopes in patients. Pairing ONM-500 adjuvant with the full-length E6 and E7 oncoproteins from human papillomavirus (HPV) cancers shows great potential to treat HPV-associated cancer in patients. Methods Based on the previously demonstrated STING-dependent T cell activation by ONM-500 [1], the nanovaccine was formulated with full-length HPV16 E6 and E7 proteins (recombinant), and the nanoparticle properties and antigen loading were characterized. In vivo lymph node accumulation following subcutaneous administration was evaluated using fluorescent nanovaccines. Direct binding of ONM-500 to recombinant human STING (CTD) was evaluated using isothermal titration calorimetry (ITC) compared to the endogenous ligand 2’,3’-cGAMP. Antitumor efficacy was evaluated in multiple syngeneic tumor models, including the TC-1 model which overexpresses HPV16 E6 and E7 with the ONM-500 vaccine in combination with anti-PD-1 checkpoint inhibitor. Long-term anti-tumor memory was evaluated in a follow-up rechallenge study after 60 days in tumor-free animals. Results Characterization of ONM-500 nanovaccine shows reproducible particle chemi-physical properties and antigen loading. The nanoparticle size substantiates the effective lymph node accumulation for antigen cross-presentation in dendritic cells following subcutaneous administration. ITC studies with human STING demonstrated effective binding by ONM-500 adjuvant. The nanovaccine anti-tumor efficacy was previously demonstrated in melanoma, colorectal, and HPV-associated syngeneic tumor models. In TC-1 tumors, ONM-500 nanovaccine containing full-length E6/E7 protein showed 100% overall survival at 55 days (figure 1). Tumor growth inhibition was also improved over E7 antigen peptide formulated nanovaccine. A rechallenge study demonstrated long-term antigen-specific anti-tumor memory response. Abstract P857 Figure 1 Conclusions ONM-500 STING-activating nanovaccines effectively deliver antigens in vivo to lymph nodes to elicit antigen-specific CTL response. The anti-tumor efficacy in multiple tumor model","PeriodicalId":16067,"journal":{"name":"Journal of Immunotherapy for Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"72611237","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
P862 Clinical benefit potentially evident with immunopharmacodynamic responses in prior-checkpoint failed metastatic melanoma patients treated with imprime PGG and pembrolizumab P862临床益处潜在明显,免疫药效学反应在先前检查点失败的转移性黑色素瘤患者中使用imprime PGG和pembrolizumab治疗
Journal of Immunotherapy for Cancer Pub Date : 2020-04-01 DOI: 10.1136/LBA2019.15
Anissa S. H. Chan, N. Bose, N. Ottoson, Xiaohong X. Qiu, B. Harrison, R. Walsh, P. Mattson, M. Gargano, J. Cox, M. Chisamore, M. Uhlik, J. Graff
{"title":"P862 Clinical benefit potentially evident with immunopharmacodynamic responses in prior-checkpoint failed metastatic melanoma patients treated with imprime PGG and pembrolizumab","authors":"Anissa S. H. Chan, N. Bose, N. Ottoson, Xiaohong X. Qiu, B. Harrison, R. Walsh, P. Mattson, M. Gargano, J. Cox, M. Chisamore, M. Uhlik, J. Graff","doi":"10.1136/LBA2019.15","DOIUrl":"https://doi.org/10.1136/LBA2019.15","url":null,"abstract":"Background Checkpoint inhibitor (CPI) monotherapy has revolutionized the treatment of melanoma, yet most patients are primary nonresponders or develop secondary resistance. Lack of antigen-specific T cell priming and/or immunosuppressive mechanisms leading to T cell exhaustion are critical cancer-extrinsic factors contributing to CPI resistance mechanisms. Immunotherapeutic agents capable of sparking de novo anti-tumor T cell responses or reinvigorating pre-existing exhausted T cell immunity could help reinstate the activity of CPI. Methods Our Phase 2, multi-center, open label study, NCT02981303 in collaboration with Merck & Co., Inc., is evaluating Imprime PGG (Imprime), a novel yeast derived, Dectin-1 agonist, β-glucan PAMP in combination with pembrolizumab (KEYTRUDA®, pembro) in heavily CPI pre-treated melanoma patients (20 patients; 65% had >2 prior CPI regimens with 17/20 having previously progressed on pembro). Patients received Imprime (4 mg/kg) + pembro (200 mg) intravenously in a 3-week cycle. Here, we present the immunopharmacodynamic (IPD) responses elicited by Imprime and pembro in the peripheral blood of 19 patients. Results In the intent-to-treat population (ITT; N=20), the disease control rate was 45% (1 CR and 8 SD), 6-month and 12-month OS rates were 65% and 45% respectively, and median OS (mOS) was 8.8 months. In the patients showing disease control, a significant increase in CH50, the classical pathway complement function (~0.7-2.6-fold), HLA-DR expression on classical monocytes (~0.61-1.94-fold) and reduction of frequency of PD-1+Tbet-EOMES+ exhausted CD8 T cells (~0.9-4-fold) was observed. Stimulation of peripheral blood mononuclear cells from a subset of patients by CD3/CD28 beads showed enhanced production of IL-2 and IFN-gamma in the CD8 T cells. Some of these IPD responses were also associated with 6-month landmark OS analyses. Additionally, whole blood gene expression analyses showed >2-fold upregulation of several myeloid and T cell activation genes including IFNg, CD83, IP-10, and IL-2RA. Enhanced OS was observed in patients with >1.3 fold increase in CH50 (8/19; HR 0.385; p=0.1) or >1.5-fold reduction in the frequency of exhausted CD8 T cells (8/19; HR 0.102; p=0.001). The IPD responses observed in the ITT population included formation of circulating immune complexes (peak levels ranging from ~4.5-16.1-fold) and production of complement activation protein SC5b9 (~3.4-25.6-fold), and increase in the frequency of HLA-DR+ myeloid cells (~0.43-3.71-fold). Conclusions Overall, these data, albeit in a small population, demonstrate that Imprime/pembro combination can drive the innate/adaptive IPD responses that are critical for providing clinical benefit to the patients who have progressed through prior CPI treatments. Ethics Approval The study was approved by central and local ethics committees depending on site requirements. The central IRB for the study is Western Institutional Review Board (WIRB), approval number 201625","PeriodicalId":16067,"journal":{"name":"Journal of Immunotherapy for Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"83404889","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 2
P864 Combination of fecal microbiota transplantation from healthy donors with anti-PD1 immunotherapy in treatment-naïve advanced or metastatic melanoma patients P864健康供体粪便微生物群移植与抗pd1免疫疗法联合治疗treatment-naïve晚期或转移性黑色素瘤患者
Journal of Immunotherapy for Cancer Pub Date : 2020-04-01 DOI: 10.1136/LBA2019.17
Saman Maleki, J. Lenehan, J. Burton, M. Silverman, S. Parvathy, Mikal El-Hajjar, M. Krishnamoorthy
{"title":"P864 Combination of fecal microbiota transplantation from healthy donors with anti-PD1 immunotherapy in treatment-naïve advanced or metastatic melanoma patients","authors":"Saman Maleki, J. Lenehan, J. Burton, M. Silverman, S. Parvathy, Mikal El-Hajjar, M. Krishnamoorthy","doi":"10.1136/LBA2019.17","DOIUrl":"https://doi.org/10.1136/LBA2019.17","url":null,"abstract":"Background Checkpoint inhibitors have changed the outcomes for patients with advanced melanoma. However, many patients still show primary resistance to single-agent therapy. Recently, the role of the gut microbiome in influencing antitumor immunity has been established. Currently, various methods of modifying the gut microbiome of cancer patients are being explored. We report the initial safety results of the first two patients treated on a phase I study combining Fecal Microbiota Transplantation (FMT) with single-agent anti-PD1 in treatment-naïve patients with advanced melanoma. Methods Two healthy donors were selected through our screening process and approximately 100 grams of fresh stool was processed and prepared for FMT as per our standardized protocol. FMT recipients were melanoma patients with unresectable or metastatic disease who were treatment naïve for their advanced disease. Bowel preparation was completed the day prior and FMT was performed using oral administration of approximately 40 capsules. Anti-PD1 was started at least 1 week after FMT to allow for microbiome engraftment. Blood and stool were analyzed at baseline (pre-FMT), before immunotherapy, and three weeks after it. Results Patient 1 was diagnosed with recurrent melanoma of the lower limb with multiple in-transit lesions refractory to control with surgery and a single intralesional injection of IL-2. Patient received stool from Donor 1 and did not experience any adverse effects from FMT. At the time of treatment #4, a solitary large cutaneous lesion stabilized but the patient experienced grade 1 diarrhoea, grade 2 nausea, and grade 2 fatigue, and grade 2 depression (NCI-CTCAE v5.0). Patient 2 was diagnosed with recurrent melanoma of the parotid gland with metastatic lesions in the lungs. Patient 2 received stool from Donor 2 and experienced only grade 1 flatus from FMT. At the time of treatment #3, the patient experienced grade 1 constipation. Both patients had a vigorous immune response to FMT measured by changes in the immune subpopulations in peripheral blood one week after FMT, including an increase in CD28+ CD8+ T cells and a decrease in PDL1+ CD3- cells. Following anti-PD1 therapy, both patients had an increase in CD39+ CD8+ T cell population. The PD1+ CD38+ CD8+ dysfunctional T cell levels decreased in both patients post-FMT and anti-PD1 therapy. Conclusions FMT combined with anti-PD1 therapy in patients with advanced melanoma appears to be safe. A measurable immune response was observed one week after FMT in both patients. One patient experienced several grade 2 toxicities with stabilization of a large cutaneous lesion. Acknowledgements This study is funded by a grant from The Lotte & John Hecht Memorial Foundation and a grant from The Medical Oncology Research Funds (MORF) from Western University. Trial Registration NCT03772899 Ethics Approval The study was approved by Western University Institutution‘s Ethics Board, approval number 113131, date of approval March","PeriodicalId":16067,"journal":{"name":"Journal of Immunotherapy for Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"82104403","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 7
P851 Identifying potential predictive biomarkers from plasma exosomes and adoptive T cells that differentiate short and long-term metastatic nasopharyngeal cancer survivors treated with chemotherapy and virus-specific T cells P851从血浆外泌体和过继T细胞中鉴定潜在的预测性生物标志物,以区分接受化疗和病毒特异性T细胞治疗的短期和长期转移性鼻咽癌幸存者
Journal of Immunotherapy for Cancer Pub Date : 2020-04-01 DOI: 10.1136/LBA2019.5
T. Shuen, Wang Who-Whong, H. Toh, Janice Lim, Cherlyn Tan, J. Kosasih, R. Cheong
{"title":"P851 Identifying potential predictive biomarkers from plasma exosomes and adoptive T cells that differentiate short and long-term metastatic nasopharyngeal cancer survivors treated with chemotherapy and virus-specific T cells","authors":"T. Shuen, Wang Who-Whong, H. Toh, Janice Lim, Cherlyn Tan, J. Kosasih, R. Cheong","doi":"10.1136/LBA2019.5","DOIUrl":"https://doi.org/10.1136/LBA2019.5","url":null,"abstract":"Background The identification of biomarkers predicting a better outcome to adoptive T cell therapy is an emerging, still nascent field. We previously completed a phase 2 clinical trial of autologous adoptive EBV-specific cytotoxic T lymphocyte (CTL) immunotherapy following gemcitabine + carboplatin chemotherapy as first line treatment in 35 advanced incurable stage 4c nasopharyngeal carcinoma (NPC) patients. Here, we aim 1) to evaluate exosome proteins for potential specific predictive biomarkers of benefit to adoptive T cell therapy and 2) to investigate if a specific immunophenotype of our generated EBV targeting CTLs correlates with survival benefit. Methods We isolated exosome from plasma samples by size exclusion chromatography and magnetic-based isolation technology, followed by fluorescence-activated cell sorting (FACS) analysis, Western blot analysis for immune-related checkpoint molecules, or mass spectrometry for exosomal peptide detection. All the generated CTLs were analysed by gene expression microarray as well as a FACS system with the use of T cell-specific 23-antibody panel for comprehensive immunophenotyping. The representative CTL samples were also subject to single-cell (sc) RNA-Seq with DNA barcoded antibodies for comprehensive integrated transcriptomics and immunophenotyping analysis. Results Patients with overall survival longer than 2 years were grouped as long survivors and the remaining patients were grouped as short survivors. Differentially expressed immune-related checkpoint molecules, such as PD1, ICOS, and CD137, were detected in in pre-treatment exosome of long and short survivors. Furthermore, more than 13,000 high confident and unique peptides which belong to 1,500 unique proteins were identified and quantified by mass spectrometry from the purified plasma exosome. Pathway enrichment analysis further showed that plasma exosome of the short survivors had significantly higher innate immune response-activating signal transduction-related peptides detected (p-value = 4.81 x 10-5). The transcriptomic analysis revealed that statistically lower expressions of SELL (CD62L) and LEF1 were found in the EBV CTL of the short survivors, suggesting that the EBV CTL of short survivors were characterized by a lesser central memory T cell phenotype. Conclusions Our data reports that specific pre-treatment plasma exosome proteins, and separately, transcriptomic profile of the generated baseline EBV CTLs prior to infusion into the patients correlate with patient survival, suggesting that they could be used together as biomarkers to predict outcome in the advanced NPC patients treated with this chemo-immunotherapy combination. More in-depth analysis of the immunophenotyping of all the CTLs and the representative CTLs’ scRNA-Seq data will be presented at the meeting. Acknowledgements This work was supported by Singapore government funding (National Medical Research Council (NMRC) – Open Fund - Large Collaborative Grant (OF-LCG) and Nat","PeriodicalId":16067,"journal":{"name":"Journal of Immunotherapy for Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"84885498","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
P853 Single cell transcriptome analysis identifies unique features in circulating CD8+ T cells that can predict immunotherapy response in melanoma patients P853单细胞转录组分析发现循环CD8+ T细胞的独特特征,可以预测黑色素瘤患者的免疫治疗反应
Journal of Immunotherapy for Cancer Pub Date : 2020-04-01 DOI: 10.1136/LBA2019.8
N. Rudqvist, R. Zappasodi, Daniel K. Wells, V. Thorsson, Alexandria P. Cogdill, A. Monette, Y. Najjar, R. Sweis, E. Wennerberg, P. Bommareddy, C. Haymaker, U. Khan, H. McGee, Wungki Park, H. Sater, C. Spencer, Nicholas P. Tschernia, M. Ascierto, Valentin V. Barsan, V. Popat, S. Valpione, B. Vincent
{"title":"P853 Single cell transcriptome analysis identifies unique features in circulating CD8+ T cells that can predict immunotherapy response in melanoma patients","authors":"N. Rudqvist, R. Zappasodi, Daniel K. Wells, V. Thorsson, Alexandria P. Cogdill, A. Monette, Y. Najjar, R. Sweis, E. Wennerberg, P. Bommareddy, C. Haymaker, U. Khan, H. McGee, Wungki Park, H. Sater, C. Spencer, Nicholas P. Tschernia, M. Ascierto, Valentin V. Barsan, V. Popat, S. Valpione, B. Vincent","doi":"10.1136/LBA2019.8","DOIUrl":"https://doi.org/10.1136/LBA2019.8","url":null,"abstract":"Background Immune checkpoint blockade (ICB) has greatly advanced the treatment of melanoma. A key component of ICB is the stimulation of CD8+ T cells in the tumor. However, ICB therapy only benefits a subset of patients and a reliable prediction method that does not require invasive biopsies is still a major challenge in the field. Methods We conducted a set of comprehensive single-cell transcriptomic analyses of CD8+ T cells in the peripheral blood (mPBL) and tumors (mTIL) from 8 patients with metastatic melanoma. Results Compared to circulating CD8+ T cells from healthy donors (hPBL), mPBLs contained subsets resembling certain features of mTIL. More importantly, three clusters (2, 6 and 15) were represented in both mPBL and mTIL. Cluster 2 was the major subset of the majority of hPBL, which phenocopied hallmark parameters of resting T cells. Cluster 6 and 15 were uniquely presented in melanoma patients. Cluster 15 had the highest PD-1 levels, with elevated markers of both activation and dysfunction/exhaustion; while Cluster 6 was enriched for ‘dormant’ cells with overall toned-down transcriptional activity except PPAR signaling, a known suppressor for T cell activation. Interestingly, unlike other mTIL clusters that would classically be defined as exhausted, Cluster 15 exhibited the highest metabolic activity (oxidative-phosphorylation and glycolysis). We further analyzed total sc-transcriptomics using cell trajectory algorithms and identified that these three clusters were the most distinct subtypes of CD8 T cells from each other, representing: resting (cluster 2), metabolically active-dysfunctional (cluster 15), and dormant phenotypes (cluster 6). Further, three unique intracellular programs in melanoma drive the transition of resting CD8+ T cells (cluster 2) to both metabolic/dysfunctional (cluster 15) and dormant states (cluster 6) that are unique to tumor bearing conditions. Based on these high-resolution analyses, we developed original algorithms to build a novel ICB response predictive model using immune-blockade co-expression gene patterns. The model was trained and tested using previously published GEO datasets containing CD8 T cells from anti-PD-1 treated patients and presented an AUC of 0.82, with 92% and 89% accuracy of ICB response in the two datasets. Conclusions We identified and analyzed unique populations of CD8+ T cells in circulation and tumor using high-resolution single-cell transcriptomics to define the landscape of CD8+ T cell states, revealing critical subsets with shared features in PBLs and TILs. Most importantly, we established an innovative model for ICB response prediction by using peripheral blood lymphocytes. Ethics Approval This study was performed under an IRB approved protocol.","PeriodicalId":16067,"journal":{"name":"Journal of Immunotherapy for Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"73072343","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
相关产品
×
本文献相关产品
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信