scholarly journals Immunohistochemical phenotyping of T cells, granulocytes, and phagocytes in the muscle of cancer patients: association with radiologically defined muscle mass and gene expression

2019 ◽  
Vol 9 (1) ◽  
Author(s):  
Ana Anoveros-Barrera ◽  
Amritpal S. Bhullar ◽  
Cynthia Stretch ◽  
Abha R. Dunichand-Hoedl ◽  
Karen J. B. Martins ◽  
...  

Abstract Background Inflammation is a recognized contributor to muscle wasting. Research in injury and myopathy suggests that interactions between the skeletal muscle and immune cells confer a pro-inflammatory environment that influences muscle loss through several mechanisms; however, this has not been explored in the cancer setting. This study investigated the local immune environment of the muscle by identifying the phenotype of immune cell populations in the muscle and their relationship to muscle mass in cancer patients. Methods Intraoperative muscle biopsies were collected from cancer patients (n = 30, 91% gastrointestinal malignancies). Muscle mass was assessed histologically (muscle fiber cross-sectional area, CSA; μm2) and radiologically (lumbar skeletal muscle index, SMI; cm2/m2 by computed tomography, CT). T cells (CD4 and CD8) and granulocytes/phagocytes (CD11b, CD14, and CD15) were assessed by immunohistochemistry. Microarray analysis was conducted in the muscle of a second cancer patient cohort. Results T cells (CD3+), granulocytes/phagocytes (CD11b+), and CD3−CD4+ cells were identified. Muscle fiber CSA (μm2) was positively correlated (Spearman’s r = > 0.45; p = < 0.05) with the total number of T cells, CD4, and CD8 T cells and granulocytes/phagocytes. In addition, patients with the smallest SMI exhibited fewer CD8 T cells within their muscle. Consistent with this, further exploration with gene correlation analyses suggests that the presence of CD8 T cells is negatively associated (Pearson’s r = ≥ 0.5; p = <0.0001) with key genes within muscle catabolic pathways for signaling (ACVR2B), ubiquitin proteasome (FOXO4, TRIM63, FBXO32, MUL1, UBC, UBB, UBE2L3), and apoptosis/autophagy (CASP8, BECN1, ATG13, SIVA1). Conclusion The skeletal muscle immune environment of cancer patients is comprised of immune cell populations from the adaptive and innate immunity. Correlations of T cells, granulocyte/phagocytes, and CD3−CD4+ cells with muscle mass measurements indicate a positive relationship between immune cell numbers and muscle mass status in cancer patients. Further exploration with gene correlation analyses suggests that the presence of CD8 T cells is negatively correlated with components of muscle catabolism.

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 34-35
Author(s):  
Anna Kalff ◽  
Sam Norton ◽  
Tiffany Khong ◽  
Malarmathy Ramachandran ◽  
Mary H. Young ◽  
...  

The LEOPARD trial evaluated lenalidomide and alternate day prednisolone (RAP) as post ASCT maintenance in newly diagnosed transplant eligible MM patients (TE NDMM). 60 patients were recruited. Estimated median potential follow-up was 44 months (IQR 26m - 52m). Median PFS from time of commencing RAP was 38.3m (95% CI, 25.8 to 54.8); median OS was not reached (71.4% of patients were alive at 36 months). Here we present the findings from correlative immune studies of this trial. Aims: To undertake mass cytometry (CyTOF) based immune profiling in patients with TE NDMM treated with RAP maintenance post ASCT. Methods: The LEOPARD trial was a phase II, multi centre, open label, single arm study of RAP maintenance after a single melphalan conditioned (200mg/m2) ASCT as part of up-front therapy. Patients were restaged at D+42 ASCT, and if eligible, were commenced on RAP maintenance (LEN 10mg daily increasing to 15mg daily after 8 weeks and alternate day prednisolone 50mg) within 8 weeks of D+0 of the ASCT. Therapy continued until toxicity/progression. CyTOF was performed in sequential samples in two selected groups of patients: long runners (LR, n=7), defined as those with PFS &gt; 36 months (median) and early relapsers (ER, n=8), defined as those who progressed/died before reaching the lower quartile of PFS. [All patients had peripheral blood collected at baseline (pre-ASCT), 6w post-ASCT and weeks 4, 8, 12, 20, 28 and 40 of RAP]. Cells were barcoded using the Cell-ID 20-Plex Pd barcoding kit (Fluidigm) followed by staining with sub-set/function defining antibodies (targeting myeloid, B, T and NK cells: CD45, CD3, CD19, CD5, CD1c, CD226. CD8, CD11c, CD16, CD127, CD138, CD123, NKG2A, TIGIT, TIM3, CD45RA. CD274, CD27, CD197, CD28, Ki67, CD66b, CD183, KLRG1, CD43, NKG2D, CD38, CD278/ICOS, CD25, HLA-DR, CD4, CD57, GramB, PD-1, CD14, CD56, CD11b, Tbet, CD33). Samples were acquired on the Helios instrument. Supervised analysis was performed to determine differences in canonical immune cell populations. Unsupervised analysis was then performed: data were clustered in the VORTEX package. Significant differences in cluster frequency were assessed by Mann-Whitney test for statistical significance. Cluster phenotypes were determined and validated via multiple visualisation approaches. Results: Median age was 56yrs for LR versus 63yrs for ER. Median PFS for LR was 46.3m (38.4 - 51.5m) versus 10.2 m (2.1 - 21.3m) for ER. Supervised analysis was performed on all samples, dichotomized into baseline and last time point sampled for each patient. At baseline, Ki67+CD8+ T cells, ICOS+CD8+ T cells, HLA-DR+CD4+ T cells and CD11c+ myeloid cells were enriched in LR compared to ER. At the last timepoint sampled, Ki67+CD8+ T cells and ICOS+CD8+ T cells were again enriched in LR compared to ER. Conversely, B-reg-like cells (CD19+CD5+CD43-) were enriched in ER compared to LR at the last timepoint sampled. Unsupervised analysis was performed on all samples (all timepoints were pooled). Five clusters were significantly enriched in LR compared to ER. Four of these clusters represented activated/cytotoxic NK cells: CD56 dim, CD16-, NKG2A(CD159a)+, NKG2D(CD314)+, Granzyme B+ and CD38+, and additional expression of CD57 on one cluster; one cluster represented a mature myeloid population, with high expression of HLA-DR, CD11b and CD11c and low expression of CD33. One cluster was significantly enriched in ER compared to LR, representing activated neutrophils, with high expression of CD66b, CD11b and CD16. The clusters that were enriched were then assessed longitudinally over all time points. There was no difference in the kinetics of these populations between groups. Conclusions Significant differences in both T-cell and NK cell populations were demonstrable at baseline in LR versus ER patients. Subsequently, durable responses to post-ASCT lenalidomide maintenance were associated with a cytotoxic, controlled immune response whereas early relapse was characterised by a more uncontrolled inflammatory response and the emergence of B-reg-like cells prior to relapse. We conclude that immune profiling at baseline and after initiation of therapy may help to predict a more sustained response to lenalidomide maintenance enabling pre-emptive tailored treatment decisions. Disclosures Kalff: Roche: Honoraria; Janssen: Honoraria; Amgen: Honoraria; CSL: Honoraria; Celgene: Honoraria. Young:Bristol Meyers Squibb: Current Employment, Current equity holder in publicly-traded company. Pierceall:Bristol Myers Squibb: Current Employment, Current equity holder in publicly-traded company. Thakurta:Bristol Myers Squibb: Current Employment, Current equity holder in publicly-traded company; Oxford University: Other: visiting professor. Oppermann:Bristol Meyers Squibb: Research Funding. Guo:Bristol Meyers Squibb: Research Funding. Reynolds:Novartis AG: Current equity holder in publicly-traded company. Spencer:AbbVie, Celgene, Haemalogix, Janssen, Sanofi, SecuraBio, Specialised Therapeutics Australia, Servier and Takeda: Consultancy; AbbVie, Amgen, Celgene, Haemalogix, Janssen, Sanofi, SecuraBio, Specialised Therapeutics Australia, Servier and Takeda: Honoraria; Amgen, Celgene, Haemalogix, Janssen, Servier and Takeda: Research Funding; Celgene, Janssen and Takeda: Speakers Bureau.


Author(s):  
Xiaofeng Yang ◽  
Tongxin Dai ◽  
Xiaobo Zhou ◽  
Hongbo Qian ◽  
Rui Guo ◽  
...  

AbstractCoronavirus disease-2019 (COVID-19), caused by SARS-CoV-2, has rapidly spread to most of countries in the world, threatening the health and lives of many people. Unfortunately, information regarding the immunological characteristics in COVID-19 patients remains limited. Here we collected the blood samples from 18 healthy donors (HD) and 38 COVID-19 patients to analyze changes in the adaptive immune cell populations and phenotypes. In comparison to HD, the lymphocyte percentage was slightly decreased, the percentages of CD4 and CD8 T cells in lymphocytes are similar, whereas B cell percentage increased in COVID-19 patients. T cells, especially CD8 T cells, showed an enhanced expression of late activation marker CD25 and exhaustion marker PD-1. Importantly, SARS-CoV-2 induced an increased percentage of T follicular helpher (Tfh)- and germinal center B-like (GCB-like) cells in the blood. However, the parameters in COVD-19 patients remained unchanged across various age groups. Therefore, we demonstrated that the T and B cells can be activated normally and exhibit functional features. These data provide a clue that the adaptive immunity in most people could be primed to induce a significant immune response against SARS-CoV-2 infection upon receiving standard medical care.


2018 ◽  
Vol 36 (6_suppl) ◽  
pp. 688-688
Author(s):  
Alejandro Sanchez ◽  
Ming Liu ◽  
Briana Nixon ◽  
Mazyar Ghanaat ◽  
Renzo DiNatale ◽  
...  

688 Background: Currently, there are no blood biomarkers that reflect the composition of immune cells in the tumor microenvironment (TME) of patients with clear cell renal cell carcinoma (ccRCC). High pre-operative neutrophil-to-lymphocyte ratio (NLR) is associated with adverse oncologic outcomes in ccRCC. However, how NLR relates to the immune cell composition in the TME is not well understood. Methods: We performed flow cytometry on tumor and adjacent normal renal tissue from a prospective cohort of patients who underwent surgery from 6/2015-7/2017. Immune cell populations, as a percent of total CD45+, were compared to baseline age, sex, body mass index (BMI), SSIGN score, sarcomatoid differentiation, and AJCC stage (I-IV). NLR was calculated using pre-operative absolute neutrophil-to-lymphocyte counts and further categorized as inflamed if NLR was ≥ 3. Correlations between continuous variables were performed using Spearman’s rank correlation. Comparisons of means were made using the Wilcoxon signed-rank test. Results: Among 48 patients, 32 (71%) were male, median age was 59 (IQR 52-66), BMI of 38.5 (IQR 26-32.6), SSIGN score of 8 (IQR 5-12), 9 (20%) had sarcomatoid differentiation, and 20 (44%) had metastases at presentation. No correlations were found between age, BMI, and different immune cell populations or NLR. A higher ratio of tissue resident (CD8a+CD49a+CD103+) to circulating (CD8a+CD49a-CD103-) CD8+ T-cells in the TME was associated with increasing stage at presentation (p = 0.02). Inflamed patients had similar total CD45+, CD8+ T cell, CD4+ T cell, and macrophage immune infiltration. However, inflamed patients had a higher proportion of CD8+ Tres infiltration (p = 0.04) and a trend towards higher neutrophil infiltration (p = 0.15). Conclusions: A higher proportion of resident CD8+T-cells correlated with advanced disease at presentation. NLR may reflect a change in the type of CD8+T cell population found in the TME. This population of CD8+ T-cells, and NLR as a possible biomarker should be validated and further investigated in a larger cohort of patients. Funding: Ruth L. Kirschstein Research Service Award T32CA082088 (A.S., M.G.)


2020 ◽  
Author(s):  
XiaoLi Wu ◽  
Hongbo Ma ◽  
YanYan Li

Abstract Abstract: Objective Gastric cancer is a malignant tumour that severely affects the health of patients. This study analyses the correlation between gastric cancer-infiltrating immune cell patterns and clinical prognosis and provides a scientific basis for the development of comprehensive tumour prevention and treatment strategies. Method Transcripts and related clinical data from 9-2019 for gastric cancer were downloaded from the TCGA database. The proportions of 22 kinds of immune cells were calculated by CIBERSORT software, and the correlation of each immune cell component ratio with tumour grade, clinical stage and overall survival (OS) was evaluated. Results A total of 413 gene transcript data sets were obtained from the TCGA database, including 381 for gastric cancer and 32 for normal tissues. The expression of various macrophages in tumour tissues was abundant. The immune cell composition, which included resting dendritic cells (p=0.02), M1 macrophages (p=0.031), resting mast cells (p=0.02), CD8 T cells (p=2.445e-04), M0 macrophages (p=6.353e-04), activated mast cells (p=0.006), neutrophils (p=0.003), resting NK cells (p=0.014), and gamma delta T cells (p=0.033), is related to the pathological grade. As the tumour stage of gastric cancer patients progresses, the proportion of some immune cells, including eosinophils (p=0.013), activated mast cells (p=0.042), neutrophils (p=0.007), and resting NK cells (p=0.036) gradually increases, while the proportion of other immune cells, for example, CD8 T cells (p=0.018), Tregs (p=0.039), M1 macrophages (p=0.018), and activated NK cells (p=0.042) gradually decreases. Higher expression of CD8 T cells suggests a better prognosis. Conclusion The composition of tumour-infiltrating immune cells differed greatly in different pathological grades and stages of gastric cancer. CD8 T cells can be used as a prognostic factor for gastric cancer patients.


2020 ◽  
Vol 38 (6_suppl) ◽  
pp. 532-532
Author(s):  
Jean-Michel Lavoie ◽  
Michael Nissen ◽  
Priya Baichoo ◽  
Lucia Nappi ◽  
Daniel Joseph Khalaf ◽  
...  

532 Background: CPIs have had a major impact on pts with mUC and mRCC. Only a small subset of pts benefit from CPI, and predictive biomarkers are needed. The role of circulating immune cells is poorly understood, but early changes after CPI exposure may predict response. We aimed to study the changes in circulating immune cell populations of pts receiving CPIs. Methods: Whole blood was collected prior to, and 3 wks after initiation of CPI in CPI-naïve pts with mUC or mRCC. PBMCs were isolated and profiled using mass cytometry (CyTOF) to provide a comprehensive overview of immune cell populations, expression of immune checkpoints, proliferation, and viability. Expression of chemokine receptors and cytokines was measured by flow cytometry. Treatment-emergent changes were correlated with response. Effects of treatment were determined by Wilcoxon signed-rank test; interactions of treatment and other variables like cluster size were determined by repeated measures two-way ANOVA. Any effects described had a significance of p<0.05. Results: Ten pts enrolled in this pilot study (mRCC = 4, mUC = 6) received anti-PDL1 (n=5), anti-PDL1/anti-CTLA4 (n=3), and anti-PDL1/chemotherapy (n=2). Best response was: 1 CR, 3 PR, 2 SD, 4 PD. Treatment induced an increase in dendritic cells (DC) and a decrease in PD1+ CD4+ and CD8+ T-cells. Elevated Ki-67, CTLA-4, LAMP-1, granzyme B and perforin expression in PD1+ cells post CPI suggested re-invigoration of exhausted T-cells. PD1+ T-cells had increased expression of the chemokine receptors CCR4 and CCR5, and decreased expression of CCR7 and CXCR4, irrespective of treatment. Response was associated with fewer CCR4+ CD4+ T cells and fewer PD1+ CD8+ T cells. Conclusions: Deep profiling by CyTOF provides a means of immune monitoring, with potential applications in clinical trials involving CPIs. Immune responses to CPIs are heterogeneous, with pt subgroups segregated by shifts in both T cell and DCs, and patterns of chemokine receptors and cytokines. Therapy reinvigorates exhausted T cells, and may cause these cells to infiltrate tumors or tumor-draining lymph nodes via chemokine receptors.


2021 ◽  
Vol 25 (Suppl 2) ◽  
pp. S96-105
Author(s):  
Jong-Kyun Lee ◽  
Yong-Seok Jee

Purpose: The purpose of this study was to elucidate the effect of resistance exercise on skeletal muscle mass-related fitness and acquired immune cell function in ovarian cancer survivors.Methods: Twelve ovarian cancer survivors aged 33–61 years participated voluntarily in this study and were divided into control group (CG, n=6) and exercise group (EG, n=6). They underwent removal of ovarian cancer and received regular care for over one year. Resistance exercise was used as the intervention program conducted 4 days a week for 12 weeks. Skeletal muscle mass, muscle strength, and endurance were assessed at baseline and at week 12. Other dependent variables included adaptive immunocytes related to helper T (Th) cells and immunosuppressors (CD4+ and CD8+).Results: After the intervention, skeletal muscle mass showed positive changes in EG com-pared to CG, although not significantly different. Muscle strength and endurance significantly increased in EG, while there was no significant change in CG. Th1, Th2, and Th1/Th2 ratio were significantly different between both groups. CD4+CD25+T cells and CD4+PD-1+T cells of EG were lower than those of CG. CD8+PD-1+T cells and CD8+TIGIT+T of EG were lower than those of CG. These results can be interpreted as the improved sensitivity of CD4+ and CD8+, which helps the secretion of myokines and cytokines, when cytotoxic substances are injected into the human body.Conclusions: This study suggests that resistance training improves upon desirable changes in adaptive immune cell responses in ovarian cancer survivors by maintaining skeletal muscle mass while developing strength and endurance.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A728-A728
Author(s):  
Yvonne Leung ◽  
Nikhil Vad ◽  
Anne Ye ◽  
Daniel Santos ◽  
Wei Cao ◽  
...  

BackgroundWe have previously demonstrated adaptive antibody responses targeting public tumor antigens in cancer patients. ATRC-101, a clinical stage, engineered version of an antibody identified in such a patient, displays robust single-agent activity in syngeneic tumor models requiring Fc receptors (FcRs) expressed by innate immune cells and the presence of CD8+ T cells. The novel target of ATRC-101 was found to be a tumor-restricted ribonucleoprotein (RNP) complex, and because RNP complexes drive T cell responses in infectious and autoimmune disease via innate immune cells, we further characterized the mechanism-of-action of ATRC-101. Here we describe changes in immune cell populations in a tumor model proximal to treatment initiation with ATRC-101.MethodsMice bearing EMT6 tumors received ATRC-101 beginning on day 7 post-tumor inoculation. Tissues were harvested between days 7 and 14 and analyzed by flow cytometry and immunohistochemistry. Transcriptome analysis was performed using RNA sequencing on whole tumors taken on days 7, 9, and 12.ResultsThe earliest significant changes induced by ATRC-101, relative to vehicle, were noted just 24 hours after dosing: increased numbers of cDC1 cells in blood, and decreased numbers of cDC2 cells in blood and M-MDSCs in tumor. A significant increase of CD8+ T cells was observed in blood 48 hours after dosing and in tumor 96 hours after dosing. Increased numbers of NK cells were also observed in blood and tumor at this later time. Multiplex analysis of circulating cytokines demonstrated a very early increase in myeloid chemo-attractants, such as MCP1 and MIP1a.Whole exome sequencing of tumor samples showed that ATRC-101 dosing drives a significant increase, relative to vehicle, in the expression of interferon-stimulated genes. Co-culturing experiments demonstrated that induced, bone marrow-derived dendritic cells are activated by ATRC-101 and its target in a dose-dependent fashion.ConclusionsDosing with ATRC-101 in the EMT6 syngeneic tumor model, in which ATRC-101 displays notable single-agent activity, leads to changes in immune cell composition in the blood and tumor, with the earliest changes observed in myeloid or myeloid-derived cell populations, and to the early appearance of myeloid chemo-attractants. We believe these data indicate that ATRC-101 acts proximally on the myeloid cell populations in the tumor, leading to a remodeling of the tumor environment and an adaptive immune response that includes CD8+ T cells driving tumor regression. Our data demonstrate that ATRC-101, bound to its target which is an RNP complex, can activate myeloid cells and are consistent with this activation occurring via FcR and Toll-like receptor (TLR) pathways.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A637-A637
Author(s):  
Manoj Chelvanambi ◽  
Ronald Fecek ◽  
Jennifer Taylor ◽  
Walter Storkus

BackgroundThe degree of immune infiltration in tumors, especially CD8+ T cells, greatly impacts patient disease course and response to interventional immunotherapy. Hence, enhancement of TIL prevalence is a preferred clinical endpoint, one that may be achieved via administration of agents that normalize the tumor vasculature (VN) leading to improved immune cell recruitment and/or that induce the development of local tertiary lymphoid structures (TLS) within the tumor microenvironment (TME).MethodsLow-dose STING agonist ADU S-100 (5 μg/mouse) was delivered intratumorally to established s.c. B16.F10 melanomas on days 10, 14 and 17 post-tumor inoculation under an IACUC-approved protocol. Treated and control, untreated tumors were isolated at various time points to assess transcriptional changes associated with VN and TLS formation via qPCR, with corollary immune cell composition changes determined using flow cytometry and immunofluorescence microscopy. In vitro assays were performed on CD11c+ BMDCs treated with 2.5 μg/mL ADU S-100 (vs PBS control) and associated transcriptional changes analyzed via qPCR or profiled using DNA microarrays. For TCRβ-CDR3 analyses, CDR3 was sequenced from gDNA isolated from enzymatically digested tumors and splenocytes.ResultsWe report that activation of STING within the TME leads to slowed melanoma growth in association with increased production of angiostatic factors including Tnfsf15 (Vegi), Cxcl10 and Angpt1, and TLS inducing factors including Ccl19, Ccl21, Lta, Ltb and Tnfsf14 (Light). Therapeutic responses from intratumoral STING activation were characterized by increased vascular normalization (VN), enhanced tumor infiltration by CD8+ T cells and CD11c+ DCs and local TLS neo-genesis, all of which were dependent on host expression of STING. Consistent with a central role for DC in TLS formation, ex vivo ADU S-100-activated mCD11c+ DCs also exhibited upregulated expression of TLS promoting factors including lymphotoxin-α (LTA), IL-36, inflammatory chemokines and type I interferons. TLS formation was associated with the development of a therapeutic TIL TCR repertoire enriched in T cell clonotypes uniquely detected within the tumor but not the peripheral circulation in support or local T cell cross-priming within the TME.ConclusionsOur data support the premise that i.t. delivery of STING agonist promotes a pro-inflammatory TME in support of VN and TLS formation, leading to the local expansion of unique TIL repertoire in association with superior anti-melanoma efficacy.


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii214-ii214
Author(s):  
Anupam Kumar ◽  
Katharine Chen ◽  
Claudia Petritsch ◽  
Theodore Nicolaides ◽  
Mariarita Santi-Vicini ◽  
...  

Abstract The determinants of the tumor-associated immune response in brain tumors are poorly understood. Using tumor samples from two molecularly distinct subtypes of lower grade glioma, MAPK-driven glioma with biallelic inactivation of CDKN2A (n=30) and IDH-mutant, 1p/19q-intact astrocytoma (n=29), we demonstrate qualitative and quantitative differences in the tumor-associated immune response and we investigate the molecular mechanisms involved. Histologically the MAPK-driven gliomas were comprised of pleomorphic xanthoastrocytoma (PXA) (n=11) and anaplastic PXA (n=19). Seven patients had paired samples from two sequential surgeries. Immune cell populations and their activity were determined by quantitative multiplex immunostaining and Digital Spatial Profiling and gene expression was analyzed by Nanostring. Functional studies were performed using established cell lines and two new patient-derived lines from MAPK-driven LGGs. MAPK-driven tumors exhibited an increased number of CD8+ T cells and tumor-associated microglial/macrophage (TAMs), including CD163+ TAMs, as compared to IDH-mutant astrocytoma. In contrast, IDH-mutant tumors had increased FOXP3+ immunosuppressive T regulatory cells. Transcriptional and protein level analyses in MAPK-driven tumors suggested an active cytotoxic T cell response with robust expression of granzyme B, present on 27% of CD8+ T cells, increased MHC class I expression, and altered cytokine profiles. Interestingly, MAPK-driven tumors also had increased expression of immunosuppressive molecules, including CXCR4, PD-L1, and VEGFA. Expression differences for cell surface and secreted proteins were confirmed in patient-derived tumor lines and functional relationships between altered chemokine expression and immune cell infiltration was investigated. Our data provide novel insights into the immune contexture of MAPK driven LGGs and suggest MAPK driven gliomas with biallelic inactivation of CDKN2A may be particularly vulnerable to immunotherapeutic modulation


Sign in / Sign up

Export Citation Format

Share Document