186 Distinct immune signatures predicting clinical response to PD-1 blockade therapy in gynecological cancers revealed by high-dimensional immune profiling

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A200-A200
Author(s):  
Yuki Muroyama ◽  
Yuki Muroyama ◽  
Sasikanth Manne ◽  
Alexandar Huang ◽  
Divij Mathew ◽  
...  

BackgroundAlthough immune checkpoint blockade revolutionized cancer therapy, response rates have been mixed in gynecological malignancies. While uterine endometrial cancer with high microsatellite instability (MSIHI) and high tumor mutational burden (TMB) respond robustly to checkpoint blockade, high-grade serous ovarian cancer (HGSOC) with low TMB respond modestly. Currently, there has been no known immune signature or T cell phenotype that predicts clinical response in gynecological tumors.MethodsTo dissect the immune landscape and T cell phenotypes in gynecological cancer patients receiving PD-1 blockade, we used high-dimensional cytometry (flow cytometry and mass cytometry (CyTOF)). We performed longitudinal deep immune profiling of PBMC from patients with recurrent uterine endometrial cancer receiving single-arm nivolumab, and HSGOC patients receiving neoadjuvant nivolumab plus platinum-based chemotherapy prior to debulking surgery.ResultsChemotherapy-resistant MSI-H uterine cancer patients treated with nivolumab had a proliferative T cell response 2–4 weeks post PD-1 blockade, consistent with responses seen in high TMB melanoma and lung cancer. The responding Ki67+ CD8 T cell population was largely CD45RAloCD27hi or CD45RAloCD27lo and highly expressed PD1, CTLA-4, and CD39, consistent with the phenotype of exhausted T cells (TEX). These exhausted-like cells are enriched in responders, whereas early expansion Tregs are enriched in non-responders. Unlike patients with uterine endometrial cancer, patients with TMBlo ovarian cancer did not have a clear proliferative CD8 T cell response after neoadjuvant nivolumab plus chemotherapy treatment, suggesting systemic immune suppression. At baseline, ovarian cancer without recurrence have more terminally differentiated effector-like CD8 T cells, and patients with recurrence have more naive-like cells. Thus, both high and low TMB gynecological tumors have distinct immune landscapes associated with clinical response. Additionally, in MSI-H uterine endometrial cancer patients, the length of time between the prior chemotherapy and the initiation of immunotherapy was negatively correlated with T cell reinvigoration post immunotherapy and clinical response. This suggests the importance of optimize therapeutic timing to maximize the therapeutic efficacy when combining immunotherapy and chemotherapy.ConclusionsCollectively, our immune profiling revealed the distinct immune signatures associated with clinical response to PD-1 blockade in gynecological cancers. Our results also suggest that TMBhi inflamed versus TMBlo cold tumor microenvironment, and timing of chemo/immunotherapy could impact differentiation and functions of T cells.Ethics ApprovalThe study was approved by MSKCC Ethics Board, approval number 17–180 and 17–182.

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1980-1980
Author(s):  
Kimberly Noonan ◽  
Lakshmi Rudraraju ◽  
Anna Ferguson ◽  
Amy Sidorski ◽  
Andrea Casildo ◽  
...  

Abstract Background Prevnar, is a multi-valent conjugate vaccine given to children and adults over 50 for the prevention of Streptococcus pneumonia, otis media and pneumococcal pneumonia. The conjugate in Prevnar is a CRM-197 protein molecule which is a nontoxic recombinant Diphtheria toxin. Prevnar serves as an excellent tool in monitoring overall immune response changes in myeloma patients’ pre and post treatment. Humoral B-cell responses can be measured by antibody responses to the pneumococcal antigens, while T cell responses to CRM-197. Clinical Study We previously conducted a study to determine the efficacy of lenalidomide to augment vaccine specific responses in patients with myeloma. Two cohorts of patients were studied. In cohort A (N=10), the first Prevnar vaccine was given two weeks prior to starting lenalidomide and the second vaccine on day 14 of cycle 2 of lenalidomide. In cohort B (N=7), both Prevnar vaccines were given on lenalidomide (day 14 of cycle 2 and 4). As we previously reported patients in cohort B had an overall better B and T cell response to Prevnar compared to cohort A. These responses were due to an overall change in B and T cell phenotype attained with lenalidomide therapy. Results Prospectively, patients in cohort B also had an unexpected overall increase in disease response and in response duration. In Cohort A only 10% of patients responded to therapy while 60% of patients in Cohort B had a clinical response. The patients with a measurable clinical response had a 5-fold increase in the percentage of tumor specific bone marrow (BM) T cells after two vaccinations with Prevnar whereas the non-responding patients had no increase in tumor specific BM T cells. Parelleling the anti-tumor response, responders showed a 15 fold increase in CRM-197 specific BM T cells after the second vaccination. Patients with no clinical response showed minimal CRM-197 T cell immunity. CRM-197 is a specific inhibitor of HB-EGF; syndecan-1 (CD138) is an HB-EGF co-receptor as well as a marker for myeloma plasma cells. We hypothesized that HB-EGF specific responses produced by vaccination with the Prevnar vaccine, and CRM-197 specifically, may have contributed to the overall increased clinical responses in our clinical trial. Responding patients had a 5-fold increase in HB-EGF specific BM T cells after vaccine 2 while clinical non-responders had no increase in HB-EGF specific BM T cells. T cells specificity for purified HB-EGF correlated with both CRM-197 and tumor specific responses. Finally the myeloma cell lines U266, H929, KMS-11 and KMS-12 co-stained for CD138 and HB-EGF with 47% of CD138+ myeloma cells co-expressing HB-EGF. Conclusions We hypothesize that the CRM-197 moiety of the Prevnar vaccine can prime T cell responses against HB-EGF on plasma cells. This immune response, in turn, weakens the tumor stromal interactions in the tumor microenvironment and potentially enhances the anti-tumor efficacy of immunomodulatory drugs such as lenalidomide. Therefore, Prevnar may possibly serve as a candidate anti-myeloma vaccine. Disclosures: No relevant conflicts of interest to declare.


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. 2607-2607
Author(s):  
David Roumanes ◽  
Evan Newell ◽  
Michael Fehlings

2607 Background: Immunotherapy recent successes have opened new avenues for the treatment of cancer and the presence of tumor-specific CD8+ T cells in tumor-bearing individuals offer a promising therapeutic target. However, the detection and profiling of such T cells are challenging due to the need to detect rare antigen-specific T cell subpopulations in patient samples that are limited in size thus making it difficult to exploit these parameters for predictive signatures of clinical response. Moreover, the identification and analysis of neoantigen-specific CD8+ T-cells in tumor-bearing individuals is challenging due to the small pool of such cells. Methods: In order to identify therapy-relevant tumor antigens and to facilitate a concurrent in-depth characterization of cells directed towards these targets, immunoSCAPE leverages the high-dimensional immune profiling capabilities of cytometry by time of flight (CyTOF) combined with a unique technology allowing the identification rare antigen-specific T-cell subsets. Results: We applied this technology to patient tumor-infiltrating lymphocytes from human cancer samples and tumor-derived neoantigens recognized by T-cells were identified and characterized. Interestingly, the majority of patient-derived tumor infiltrates consisted of tumor-unrelated T-cells characterized by a diverse phenotype. Strikingly, the expression of CD39 was absent from these bystander cells, suggesting that CD39 could be a useful biomarker for the identification of putative tumor-reactive T cells. Conclusions: Simultaneous immune profiling revealed that tumor-unrelated, bystander CD8+ T-cells are phenotypically different in human tumor infiltrates and identified CD39 as a putative marker of neoantigen-specific T-cells. By providing insights into the nature, frequency and phenotype of antigen-specific T-cells, immunoSCAPE’s unique target discovery and high-dimensional immune profiling platform is a valuable tool for the development of novel diagnostic and therapeutic strategies in immunotherapy.


2018 ◽  
Vol 2018 ◽  
pp. 1-7 ◽  
Author(s):  
Hani Choudhry ◽  
Nawal Helmi ◽  
Wesam H. Abdulaal ◽  
Mustafa Zeyadi ◽  
Mazin A. Zamzami ◽  
...  

IL-2 is a powerful immune growth factor and it plays important role in sustaining T cell response. The potential of IL-2 in expanding T cells without loss of functionality has led to its early use in cancer immunotherapy. IL-2 has been reported to induce complete and durable regressions in cancer patients but immune related adverse effects have been reported (irAE). The present review discusses the prospects of IL-2 in immunotherapy for cancer.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A289-A290
Author(s):  
Myles Dillon ◽  
Lianjie Li ◽  
Jeongsook Bang ◽  
Nicholas Gaspar ◽  
Jessica Kuhnert ◽  
...  

BackgroundTo better understand the immune stimulatory mechanisms of Oncolytic virus (OV), we evaluated the circulating OV-specific T cell response in patients during the course of OV therapy. Patients with histologically confirmed advanced clear cell renal cell carcinoma, who were naïve or refractory to prior systemic treatment and who had no prior treatment with immune checkpoint inhibitors, were treated with 4 weekly intravenous infusions of Pexa-Vec at 1 × 109 plaque forming units starting at Day -7 plus Cemiplimab (350 mg every 3 weeks) from Day 1. Radiographic assessments per RECIST 1.1 were performed centrally every 9 weeks from Day 1. Peripheral blood mononuclear cells (PBMCs) were collected and cryopreserved at baseline and 29 days post initial Pexa-Vec treatment.MethodsWe performed functional IFNγ ELISPOT analysis on longitudinal PBMC samples using a custom panel of OV epitopes and culture conditions designed to measure existing OV-specific memory T cell cytolytic activity [1]. PBMC samples were tested for IFNγ release following stimulation with OV peptides using two different assay conditions: 1) measurement following direct ex vivo stimulation with OV peptides alone, and 2) measurement following 10 days of T cell expansion in the presence of OV peptides, T cell supportive cytokines (GM-CSF, IL-4, IL-7 and IL-15), and autologous dendritic cells. The number of OV-specific IFNγ spots was correlated with the clinical response and tumor regression.ResultsIn preliminary analyses, 8 of the 11 (72.7%) patients showed tumor burden reduction, 4 of whom had ≥30% confirmed reduction that qualify as RECIST1.1 PRs (figure 1 and 2). OV-specific IFNγ+ T cells were detected in only 3 out of 11 patients in the non-expanded ELISPOT culture conditions (figure 3A), but in 8 out of 11 patients when T cells were first expanded for 10 days in the presence of OV peptides prior to ELISPOT, which trended toward a correlation with the preliminary clinical response assessment (figure 3B). Prolonged stimulation with CMV, EBV and Influenza peptides did not show any correlation (R2 = 0.005), suggesting that the treatment and culture expansion influenced relevant OV-specific memory T cell proliferation.Abstract 264 figure 1Tumor response and% change in tumor burdenPatients received 6 to 15 rounds of Cemiplimab at the time of data collection.*One patient who had Progressive Disease experienced significant regression of primary tumor but developed metastatic lesions.Abstract 264 Figure 2Tumor volume change in individual patientsPercentage of tumor volume change in individual patients over the course of treatment.Abstract 264 Figure 3Correlation between ELISPOT and tumor responseCorrelation between T-cell response (ELISPOT) and tumor volume change.PBMC sample detection range: 9-fold inscrease in the detection range of IFNγ producing cells up to 1000 SFC/2 × 105 cells, (3B) from a detection range of fewer than 110 SFC/2 × 105 cells (3A).Pre-expanded ELISPOT revealed a trend of correlation between CD8+ T cell response and tumor volume reduction even at an early sample collection time point (Day 29) with R2=0.3031 and p-value = 0.0793.ConclusionsThese results suggest that OV-specific T cell responses can be induced by OV therapy. In addition, 10-day expansion of low levels of OV-specific circulating T cells can amplify signals in ELISPOT analysis and might enable systemic tracking of patient responses in blood samples collected at early time points. The observed CD8+ T cell response to oncolytic vaccinia virus in patients supports the rationale for combination treatment with Pexa-Vec and immune checkpoint inhibitors.AcknowledgementsSun Young Rha, Yonsei Cancer Center, Severance Hospital, Yonsei University Health System, Seoul, Republic of KoreaJamie Merchan, University of Miami Health System, Miami, FL, USASung Yong Oh, Dong-A University Hospital, Busan, Republic of KoreaChan Kim, Cha University Bundang Medical Center, Seongnam, Republic of KoreaWoo Kyun Bae, Chonnam National University Hwasun Hospital, Hwasun, Republic of KoreaHyun Woo Lee, Ajou University Hospital, Suwon, Republic of Korea.Trial RegistrationNCT03294083Ethics ApprovalThe study was approved by University of Miami Institutional Reivew Board, approval number 20180055.ReferenceDillon M, Jeong S, De Silva N, et al. Tracking oncolytic virus-specific CD8+ T cells with epitope-based, HLA-agnostic peptides in a renal cell carcinoma clinical trial. CRI-CIMT-EATI-AACR INTERNATIONAL CANCER IMMUNOTHERAPY CONFERENCE 2019. Abstract #A067


2019 ◽  
Vol 8 (3) ◽  
pp. 380 ◽  
Author(s):  
Chiara Napoletano ◽  
Ilary Ruscito ◽  
Filippo Bellati ◽  
Ilaria Grazia Zizzari ◽  
Hassan Rahimi ◽  
...  

Increasing evidence strongly suggests that bevacizumab compound impacts the immunological signature of cancer patients and normalizes tumor vasculature. This study aims to investigate the correlation between the clinical response to bevacizumab-based chemotherapy and the improvement of immune fitness of multi-treated ovarian cancer patients. Peripheral blood mononuclear cells (PBMCs) of 20 consecutive recurrent ovarian cancer patients retrospectively selected to have received bevacizumab or non-bevacizumab-based chemotherapy (Bev group and Ctrl group, respectively) were analyzed. CD4, CD8, and regulatory T cell (Treg) subsets were monitored at the beginning (T0) and after three and six cycles of treatment, together with IL10 production. A lower activated and resting Treg subset was found in the Bev group compared with the Ctrl group until the third therapy cycle, suggesting a reduced immunosuppressive signature. Indeed, clinically responding patients in the Bev group showed a high percentage of non-suppressive Treg and a significant lower IL10 production compared with non-responding patients in the Bev group after three cycles. Furthermore, clinically responding patients showed a discrete population of effector T cell at T0 independent of the therapeutic regimen. This subset was maintained throughout the therapy in only the Bev group. This study evidences that bevacizumab could affect the clinical response of cancer patients, reducing the percentage of Treg and sustaining the circulation of the effector T cells. Results also provide a first rationale regarding the positive immunologic synergism of combining bevacizumab with immunotherapy in multi-treated ovarian cancer patients.


2020 ◽  
Vol 8 (1) ◽  
pp. e000560 ◽  
Author(s):  
Victoria A Brentville ◽  
Rachael L Metheringham ◽  
Ian Daniels ◽  
Suha Atabani ◽  
Peter Symonds ◽  
...  

BackgroundStress-induced post-translational modifications occur during autophagy and can result in generation of new epitopes and immune recognition. One such modification is the conversion of arginine to citrulline by peptidylarginine deiminase enzymes.MethodsWe used Human leukocyte antigen (HLA) transgenic mouse models to assess the immunogenicity of citrullinated peptide vaccine by cytokine Enzyme linked immunosorbant spot (ELISpot) assay. Vaccine efficacy was assessed in tumor therapy studies using HLA-matched B16 melanoma and ID8 ovarian models expressing either constitutive or interferon-gamma (IFNγ) inducible Major Histocompatibility Complex (MHC) class II (MHC-II) as represented by most human tumors. To determine the importance of CD4 T cells in tumor therapy, we analyzed the immune cell infiltrate into murine tumors using flow cytometry and performed therapy studies in the presence of CD4 and CD8 T cell depletion. We assessed the T cell repertoire to citrullinated peptides in ovarian cancer patients and healthy donors using flow cytometry.ResultsThe combination of citrullinated vimentin and enolase peptides (Modi-1) stimulated strong CD4 T cell responses in mice. Responses resulted in a potent anti-tumor therapy against established tumors and generated immunological memory which protected against tumor rechallenge. Depletion of CD4, but not CD8 T cells, abrogated the primary anti-tumor response as well as the memory response to tumor rechallenge. This was further reinforced by successful tumor regression being associated with an increase in tumor-infiltrating CD4 T cells and a reduction in tumor-associated myeloid suppressor cells. The anti-tumor response also relied on direct CD4 T cell recognition as only tumors expressing MHC-II were rejected. A comparison of different Toll-like receptor (TLR)-stimulating adjuvants showed that Modi-1 induced strong Th1 responses when combined with granulocyte-macrophage colony-stimulating factor (GMCSF), TLR9/TLR4, TLR9, TLR3, TLR1/2 and TLR7 agonists. Direct linkage of the TLR1/2 agonist to the peptides allowed the vaccine dose to be reduced by 10-fold to 100-fold without loss of anti-tumor activity. Furthermore, a CD4 Th1 response to the citrullinated peptides was seen in ovarian cancer patients.ConclusionsModi-1 citrullinated peptide vaccine induces potent CD4-mediated anti-tumor responses in mouse models and a CD4 T cell repertoire is present in ovarian cancer patients to the citrullinated peptides suggesting that Modi-1 could be an effective vaccine for ovarian cancer patients.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A532-A532
Author(s):  
Casey Ager ◽  
Aleksander Obradovic ◽  
Juan Arriaga ◽  
Matthew Chaimowitz ◽  
Cory Abate-Shen ◽  
...  

BackgroundThe theory of cancer immunoediting, which describes the dynamic interactions between tumors and host immune cells that shape the character of each compartment, is foundational for understanding cancer immunotherapy. Few models exist that facilitate in-depth study of each of the three canonical phases of immunoediting: elimination, equilibrium, and escape. Here, we perform high dimensional longitudinal immune profiling of NPK-C1, a transplantable prostate tumor model that recapitulates the three phases of immunoediting spontaneously in immunocompetent C57BL/6 animals.MethodsWe generated a 28-color immune phenotyping panel to interrogate the NPK-C1 microenvironment using a Cytek Aurora spectral flow cytometer. We analyzed NPK-C1 tumors on days 10, 15, 20 and 24 post-implantation, representing elimination, equilibrium, early escape, and late escape phases, respectively. These data were analyzed by both traditional gating and with an optimized dimensionality reduction and unsupervised clustering workflow. We additionally performed in vivo depletion studies of T cell and granulocyte subsets at early and late time points to determine if these bulk populations are required for immunoediting during elimination and equilibrium/escape.ResultsWe found that a cluster of activated CD4 effector T cells were enriched early during elimination phase and were overrepresented in NPK-C1 tumors which regress rather than progress to escape. CD4 in vivo depletion studies validated a functional role for CD4 T cells in suppressing NPK-C1 progression at these phases. Additionally, a central memory-like cytotoxic CD8 T cell cluster was enriched in regressing NPK-C1 tumors, and CD8 depletion allowed NPK-C1 progression throughout immunoediting. Regulatory T cells (Tregs) as a whole were counterintuitively enriched in regressing tumors, however high dimensional analysis revealed their significant phenotypic diversity, with a number of Treg subpopulations enriched in progressing tumors. In the myeloid compartment, we found that iNOS+ DC-like cells were enriched in regressing tumors, while CD103+ DCs were counterintuitively associated with late stage tumor progression.ConclusionsThese data introduce a new model – NPK-C1 – to study immunoediting and suggest both CD8 and CD4 T cells are required to suppress tumor outgrowth throughout each phase of cancer immunoediting, while myeloid populations exhibit significant phenotypic and functional diversity throughout this process. Further, our identification of unique sub-populations of myeloid and T cells correlating with either regression or progression to escape highlights a role for multi-dimensional flow-based analyses to more deeply understand immune cell dynamics in the tumor microenvironment.Ethics ApprovalAll experiments and procedures for this study were approved by the Columbia University Medical Center Institutional Animal Care and Use Committee (IACUC)


Sign in / Sign up

Export Citation Format

Share Document