Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Front Immunol ; 13: 1074740, 2022.
Article in English | MEDLINE | ID: mdl-36601119

ABSTRACT

Access to commercial CD19 CAR-T cells remains limited even in wealthy countries like Canada due to clinical, logistical, and financial barriers related to centrally manufactured products. We created a non-commercial academic platform for end-to-end manufacturing of CAR-T cells within Canada's publicly funded healthcare system. We report initial results from a single-arm, open-label study to determine the safety and efficacy of in-house manufactured CD19 CAR-T cells (entitled CLIC-1901) in participants with relapsed/refractory CD19 positive hematologic malignancies. Using a GMP compliant semi-automated, closed process on the Miltenyi Prodigy, T cells were transduced with lentiviral vector bearing a 4-1BB anti-CD19 CAR transgene and expanded. Participants underwent lymphodepletion with fludarabine and cyclophosphamide, followed by infusion of non-cryopreserved CAR-T cells. Thirty participants with non-Hodgkin's lymphoma (n=25) or acute lymphoblastic leukemia (n=5) were infused with CLIC-1901: 21 males (70%), median age 66 (range 18-75). Time from enrollment to CLIC-1901 infusion was a median of 20 days (range 15-48). The median CLIC-1901 dose infused was 2.3 × 106 CAR-T cells/kg (range 0.13-3.6 × 106/kg). Toxicity included ≥ grade 3 cytokine release syndrome (n=2) and neurotoxicity (n=1). Median follow-up was 6.5 months. Overall response rate at day 28 was 76.7%. Median progression-free and overall survival was 6 months (95%CI 3-not estimable) and 11 months (95% 6.6-not estimable), respectively. This is the first trial of in-house manufactured CAR-T cells in Canada and demonstrates that administering fresh CLIC-1901 product is fast, safe, and efficacious. Our experience may provide helpful guidance for other jurisdictions seeking to create feasible and sustainable CAR-T cell programs in research-oriented yet resource-constrained settings. Clinical trial registration: https://clinicaltrials.gov/ct2/show/NCT03765177, identifier NCT03765177.


Subject(s)
Hematologic Neoplasms , Lymphoma, Non-Hodgkin , Male , Humans , Aged , T-Lymphocytes , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Cyclophosphamide , Hematologic Neoplasms/therapy , Recurrence , Antigens, CD19
2.
Clin Cancer Res ; 27(14): 4089-4100, 2021 07 15.
Article in English | MEDLINE | ID: mdl-33963000

ABSTRACT

PURPOSE: Tumor-infiltrating lymphocytes (TIL) are strongly associated with survival in most cancers; however, the tumor-reactive subset that drives this prognostic effect remains poorly defined. CD39, CD103, and PD-1 have been independently proposed as markers of tumor-reactive CD8+ TIL in various cancers. We evaluated the phenotype, clonality, and prognostic significance of TIL expressing various combinations of these markers in high-grade serous ovarian cancer (HGSC), a malignancy in need of more effective immunotherapeutic approaches. EXPERIMENTAL DESIGN: Expression of CD39, CD103, PD-1, and other immune markers was assessed by high-dimensional flow cytometry, single-cell sequencing, and multiplex immunofluorescence of primary and matched pre/post-chemotherapy HGSC specimens. RESULTS: Coexpression of CD39, CD103, and PD-1 ("triple-positive" phenotype) demarcated subsets of CD8+ TIL and CD4+ regulatory T cells (Treg) with a highly activated/exhausted phenotype. Triple-positive CD8+ TIL exhibited reduced T-cell receptor (TCR) diversity and expressed genes involved in both cytolytic and humoral immunity. Triple-positive Tregs exhibited higher TCR diversity and a tumor-resident phenotype. Triple-positive TIL showed superior prognostic impact relative to TIL expressing other combinations of these markers. TIGIT was uniquely upregulated on triple-positive CD8+ effector cells relative to their CD4+ Treg counterparts. CONCLUSIONS: Coexpression of CD39, CD103, and PD-1 demarcates highly activated CD8+ and CD4+ TIL with inferred roles in cytolytic, humoral, and regulatory immune functions. Triple-positive TIL demonstrate exceptional prognostic significance and express compelling targets for combination immunotherapy, including PD-1, CD39, and TIGIT.


Subject(s)
Cystadenocarcinoma, Serous/immunology , Cystadenocarcinoma, Serous/pathology , Lymphocytes, Tumor-Infiltrating , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Antigens, CD/biosynthesis , Apyrase/biosynthesis , Cystadenocarcinoma, Serous/metabolism , Female , Humans , Integrin alpha Chains/biosynthesis , Lymphocytes, Tumor-Infiltrating/metabolism , Ovarian Neoplasms/metabolism , Prognosis , Programmed Cell Death 1 Receptor/biosynthesis
3.
Gynecol Oncol ; 158(1): 167-177, 2020 07.
Article in English | MEDLINE | ID: mdl-32446718

ABSTRACT

OBJECTIVE: We recently showed that tumors with an immunologically 'cold' phenotype are enriched for expression of stemness-associated genes and PVR/CD155, the ligand of the immunosuppressive molecule TIGIT. To explore the therapeutic implications of this finding, we investigated the relationship between PVR/CD155 expression, tumor-infiltrating lymphocytes (TIL), and prognosis in high-grade serous ovarian cancer (HGSC) and other cancers. METHODS: Expression of CD155, TIGIT, PD-1, PD-L1, and other immune markers in HGSC was assessed by high-dimensional flow cytometry, multi-color histological imaging, and/or gene expression profiling. The prognostic significance of PVR/CD155 and CD274/PD-L1 expression was assessed bioinformatically in HGSC and 32 other cancers in The Cancer Genome Atlas. RESULTS: T cells from HGSC frequently co-expressed TIGIT and PD-1, and the ratio of TIGIT to PD-1 expression increased markedly after in vitro expansion with a clinically relevant protocol. CD155 was commonly expressed on malignant epithelium in HGSC and showed a negative or non-significant association with TIL. In contrast, PD-L1 was predominantly expressed by tumor-associated macrophages and positively associated with TIL. These contrasts between CD155 and PD-L1 were seen across HGSC patients, across metastatic sites within individual patients, and even within individual tumor deposits. PVR/CD155 and CD274/PD-L1 exhibited divergent prognostic associations across diverse cancer types in TCGA, including HGSC. CONCLUSIONS: CD155 and PD-L1 exhibit contrasting expression patterns, TIL associations and prognostic significance, suggesting they represent non-redundant immunosuppressive mechanisms. The CD155/TIGIT pathway represents a compelling immunotherapeutic target for HGSC and for immunologically cold tumors in general.


Subject(s)
B7-H1 Antigen/biosynthesis , B7-H1 Antigen/immunology , Cystadenocarcinoma, Serous/immunology , Neoplasms/immunology , Ovarian Neoplasms/immunology , Receptors, Virus/biosynthesis , Receptors, Virus/immunology , Aged , B7-H1 Antigen/genetics , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Female , Gene Expression , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Middle Aged , Neoplasm Grading , Neoplasm Staging , Neoplasms/genetics , Neoplasms/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Programmed Cell Death 1 Receptor/biosynthesis , Programmed Cell Death 1 Receptor/immunology , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/immunology , Receptors, Virus/genetics
4.
Mol Ther Methods Clin Dev ; 17: 393-399, 2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32128343

ABSTRACT

Mycoplasma species (spp.) bacteria can infect cell cultures, posing a potential threat to recipients of cell therapy products. Conventional Mycoplasma testing methods are highly sensitive but typically require a minimum of 28 days to produce results. This delay is problematic if rapid results are needed to inform treatment decisions. Nucleic acid amplification technique (NAT) methods have been gaining favor for Mycoplasma testing due to their speed and specificity; however, they must first be qualified as meeting or exceeding the sensitivity of the compendial method. We present herein a NAT method for the detection of Mycoplasma that circumvents the need for live Mycoplasma spp. in the test procedure by instead being qualified using Mycoplasma spp. genomic DNA. We have demonstrated a lower limit of detection that exceeds the regulatory requirements set by Health Canada. This assay is now being used to screen clinical cell therapy products manufactured at our center.

5.
PLoS One ; 14(1): e0210902, 2019.
Article in English | MEDLINE | ID: mdl-30657796

ABSTRACT

Extracellular bacteria that spread via the vasculature employ invasive mechanisms that mirror those of metastatic tumor cells, including intravasation into the bloodstream and survival during hematogenous dissemination, arrestation despite blood flow, and extravasation into distant tissue sites. Several invasive bacteria have been shown to exploit normal platelet function during infection. Due to their inherent ability to interact with and influence other cell types, platelets play a critical role in alteration of endothelial barrier permeability, and their role in cancer metastasis has been well established. The highly invasive bacterium and causative agent of syphilis, Treponema pallidum subspecies pallidum, readily crosses the endothelial, blood-brain and placental barriers. However, the mechanisms underlying this unusual and important aspect of T. pallidum pathogenesis are incompletely understood. In this study we use darkfield microscopy in combination with flow cytometry to establish that T. pallidum interacts with platelets. We also investigate the dynamics of this interaction and show T. pallidum is able to activate platelets and preferentially interacts with activated platelets. Platelet-interacting treponemes consistently exhibit altered kinematic (movement) parameters compared to free treponemes, and T. pallidum-platelet interactions are reversible. This study provides insight into host cell interactions at play during T. pallidum infection and suggests that T. pallidum may exploit platelet function to aid in establishment of disseminated infection.


Subject(s)
Blood Platelets/microbiology , Host Microbial Interactions/physiology , Syphilis/blood , Syphilis/microbiology , Treponema pallidum/pathogenicity , Humans , In Vitro Techniques , Microscopy, Video , Movement , Platelet Activation , Rotation , Treponema pallidum/physiology
6.
Front Immunol ; 9: 1741, 2018.
Article in English | MEDLINE | ID: mdl-30093907

ABSTRACT

Resident memory T cells (TRM) are a recently identified subset of long-lived memory T cells that are characterized in terms of their unique surface phenotype combined with a non-recirculating pattern of localization to non-lymphoid, peripheral tissues. TRM have quickly become a key area of focus in understanding immune responses to microbial infection in so-called "barrier" tissues, and appear to be particularly critical for protection against repeat exposure at the same site. More recently, tumor-infiltrating T cells with canonical TRM features are being identified in human cancers, in particular cancers of epithelial origin, and their presence is broadly found to be associated with favorable long-term prognosis. Moreover, recent studies have shown that these "resident memory-like" tumor-infiltrating lymphocytes (referred to herein as TILRM) are uniquely activated in melanoma patients undergoing PD-1 directed checkpoint blockade therapy. Accordingly, there is much interest at present regarding the biology of these cells and their precise role in anti-cancer immunity. Herein, we review the current state of the literature regarding TILRM with a specific emphasis on their specificity, origins, and relationship to conventional pathogen-specific TRM and speculate upon the way(s) in which they might contribute to improved prognosis for cancer patients. We discuss the growing body of evidence that suggests TILRM may represent a population of bona-fide tumor-reactive T cells and the attractive possibility of leveraging this cell population for future immunotherapy.


Subject(s)
Immunologic Memory , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/immunology , Animals , Antigens, CD/metabolism , Biomarkers , Humans , Integrin alpha Chains/metabolism , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Lymphocyte Subsets/pathology , Lymphocytes, Tumor-Infiltrating/pathology , Neoplasms/metabolism , Neoplasms/mortality , Neoplasms/pathology , Phenotype , Prognosis , Sex Factors , T-Cell Antigen Receptor Specificity , Tumor Microenvironment/immunology
7.
Oncotarget ; 8(31): 51641-51651, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28881675

ABSTRACT

PURPOSE: PD-L1 is thought to play an important role in the antitumor immune response. In this study, we investigated the expression of PD-L1 within breast tumor subsets to better define its prognostic significance. METHODS: Immunohistochemistry was performed to determine PD-L1 tumor cell expression and to enumerate CD8, CD4 and CD68 tumor-infiltrating leucocytes (TIL) in a cohort of 443 breast cancers categorized by molecular subtype. RESULTS: Across the entire cohort, PD-L1 tumor cell expression was observed in 73/443 (16.5%) cases and associated with known indicators of poor prognosis, including low patient age, high tumor grade, ER/PR negative status, but not with outcome. However, in the Triple Negative breast cancer subset PD-L1 was associated with better recurrence free survival (RFS) especially within the Basal-like subset (Hazard ratio = 0.39, 95% CI = 0.22 - 0.86, p = 0.018). Combined PD-L1/epithelial CD8 positive status was also strongly associated with better RFS and OS (Hazard ratio = 0.12, 95% CI = 0.10 - 0.71, p = 0.010 and Hazard ratio = 0.11, 95% CI = 0.11 - 0.68, p = 0.006 respectively) in the Basal-like subgroup. CONCLUSIONS: PD-L1 expression is associated with better patient survival in Basal-like breast cancer.

8.
Oncoimmunology ; 6(7): e1321184, 2017.
Article in English | MEDLINE | ID: mdl-28811957

ABSTRACT

Oncogenic "driver" mutations are theoretically attractive targets for the immunotherapy of lymphoid cancers, yet the proportion that can be recognized by T cells remains poorly defined. To address this issue without any confounding effects of the patient's immune system, we assessed T cells from 19 healthy donors for recognition of three common driver mutations in lymphoma: MYD88L265P, EZH2Y641F , and EZH2Y641N . Donors collectively expressed the 10 most prevalent HLA class I alleles, including HLA-A*02:01. Peripheral blood T cells were primed with peptide-loaded dendritic cells (DC), and reactive T cells were assessed for recognition of naturally processed mutant versus wild type full-length proteins. After screening three driver mutations across 17-26 HLA class I alleles and 3 × 106-3 × 107 T cells per donor, we identified CD4+ T cells against EFISENCGEII from EZH2Y641N (presented by HLA-DRB1*13:02) and CD8+ T cells against RPIPIKYKA from MYD88L265P (presented by HLA-B*07:02). We failed to detect RPIPIKYKA-specific T cells in seven other HLA-B*07:02-positive donors, including two lymphoma patients. Thus, healthy donors harbor T cells specific for common driver mutations in lymphoma. However, such responses appear to be rare due to the combined limitations of antigen processing, HLA restriction, and T cell repertoire size, highlighting the need for highly individualized approaches for selecting targets.

9.
Oncotarget ; 8(8): 12664-12674, 2017 Feb 21.
Article in English | MEDLINE | ID: mdl-27058619

ABSTRACT

CD74 (invariant chain) plays a role in MHC class II antigen presentation. We assessed CD74 and MHCII expression in tumor cells, as well as CD8, CD4, and CD68 tumor infiltrating leucocyte (TIL) density by immunohistochemistry in a cohort of 492 breast cancer patients. CD74 expression was associated with poor prognostic markers including patient age, tumor grade, ER status, non-Luminal A subtypes, and with MHCII expression and higher TIL densities, particularly in the Basal-like subgroup. Univariate analysis showed a favorable prognostic effect of CD74 (Hazard ratio = 0.46, 95% CI = 0.26-0.89, p = 0.022) and for combined CD74/MHCII (Hazard ratio = 0.26, 95% CI = 0.17-0.81, p = 0.014) positive status for overall survival that was only manifested in the Basal-like subgroup. CD74 and MHCII expression is associated with patient survival in Basal-like breast cancer, and the association with TIL may reflect an effective intratumoral immune response.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/biosynthesis , Biomarkers, Tumor/analysis , Breast Neoplasms/immunology , Histocompatibility Antigens Class II/biosynthesis , Lymphocytes, Tumor-Infiltrating/immunology , Adult , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Disease-Free Survival , Female , Histocompatibility Antigens Class II/immunology , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Middle Aged , Prognosis , Tissue Array Analysis
10.
Clin Cancer Res ; 22(24): 6290-6297, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27267849

ABSTRACT

PURPOSE: CD103 is expressed in several immune cell types but in the context of the intratumoral immune response may be most important as a marker of antigen-activated CD8 T cells. METHODS: We have examined the prognostic significance of CD103 TILs in breast cancer by IHC in a cohort of 424 breast cancer patients. RESULTS: CD103 TILs were present in all subtypes but were more abundant in ER-negative tumors where CD103 TILs were preferentially localized to the intraepithelial compartment. CD103 was associated with tumor size, tumor grade, and ER/PR status (P < 0.05). CD103 TIL density and the epithelial to stromal ratio was highest in the basal-like tumors. Intraepithelial CD103 but not intrastromal CD103 was associated with better relapse-free and overall survival in basal-like subtype tumors [HR = 0.28; 95% confidence interval (CI), 0.17-0.72; P = 0.0047 and HR = 0.25; 95% CI, 0.17-0.66; P = 0.0017, respectively). CD8 status showed similar but less significant associations, but the combination of dual CD103+CD8+ TIL status was the most strongly prognostic combination for relapse-free and overall survival (HR = 0.10; 95% CI, 0.07-0.62; P = 0.006 and HR = 0.09; 95% CI, 0.07-0.57; P = 0.003, respectively). CONCLUSIONS: CD103 TILs are indicative of a good prognosis specifically within the basal-like subtype of breast cancer. Clin Cancer Res; 22(24); 6290-7. ©2016 AACR.


Subject(s)
Antigens, CD/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Integrin alpha Chains/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Adult , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cohort Studies , Disease-Free Survival , Female , Humans , Lymphocytes, Tumor-Infiltrating/pathology , Prognosis
11.
Gynecol Oncol ; 141(2): 293-302, 2016 05.
Article in English | MEDLINE | ID: mdl-26972336

ABSTRACT

OBJECTIVE: As a negative regulator of T cells, Programmed Death Ligand 1 (PD-L1) is both an indicator and inhibitor of anti-tumor immune responses, which has led to confusion about its prognostic significance. We investigated the primary source of PD-L1 expression in epithelial ovarian cancer and its relationship to tumor-infiltrating lymphocytes (TIL) and associated gene products. METHODS: Tissue microarrays containing high-grade serous carcinomas (HGSC) and endometrioid, clear cell and mucinous ovarian cancers from optimally debulked patients were assessed by immunohistochemistry for expression of PD-L1 and other markers (CD68, CD3, CD8, PD-1, CD103, FoxP3 and CD25). The Cancer Genome Atlas was interrogated for associations between PD-L1 expression and immune-related transcriptional and genomic features of HGSC. RESULTS: PD-L1 was primarily expressed by tumor-associated CD68(+) macrophages rather than tumor cells. PD-L1(+) cells frequently co-localized with CD8, CD4 and PD-1(+) TIL, CD25(+)FoxP3(+) Tregs, and other TIL subsets. PD-L1(+) cells were prognostically favorable in HGSC. Moreover, the presence of both PD-L1(+) cells and CD8 TIL was associated with better prognosis than CD8 TIL alone. PD-L1 gene expression was independent of BRCA status. At the transcriptional level, PD-L1 was associated with both cytolytic (granzyme B, T-bet and IFN-γ) and suppressive (PD-1, CTLA-4, LAG3 and IDO-1) gene products. CONCLUSIONS: PD-L1 is primarily expressed by macrophages in ovarian cancer and is strongly associated with both cytolytic and regulatory TIL subsets, resulting in a net positive association with survival. Tumors containing PD-L1(+) macrophages appear caught in an immunological stalemate that may require multi-pronged immunotherapy to alleviate.


Subject(s)
B7-H1 Antigen/biosynthesis , Cystadenocarcinoma, Serous/immunology , Cystadenocarcinoma, Serous/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms, Glandular and Epithelial/immunology , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , B7-H1 Antigen/immunology , Carcinoma, Ovarian Epithelial , Cystadenocarcinoma, Serous/pathology , Female , Humans , Immunohistochemistry , Lymphocytes, Tumor-Infiltrating/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Prognosis , Tissue Array Analysis
12.
Cancer Immunol Res ; 3(8): 926-35, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25957117

ABSTRACT

αE(CD103)ß7 is a TGFß-regulated integrin that mediates retention of lymphocytes in peripheral tissues by binding to E-cadherin expressed on epithelial cells. We recently reported that αE(CD103)ß7 specifically demarcates intraepithelial CD8(+) tumor-infiltrating lymphocytes (CD8 TIL) in ovarian cancer and that CD103(+) TIL have a surface profile consistent with an active effector phenotype (HLA-DR(+), Ki67(+), and CD127(lo)). These findings led us to hypothesize that, over time, CD103-mediated retention of CD8 TIL within the tumor epithelium might result in chronic stimulation by tumor antigen, which in turn might lead to an exhausted phenotype. To investigate this possibility, we evaluated PD-1 expression in a large cohort of ovarian tumors (N = 489) with known CD103(+) TIL content. PD-1(+) cells were present in 38.5% of high-grade serous carcinomas (HGSC), but were less prevalent in other histologic subtypes. PD-1(+) TIL were strongly associated with increased disease-specific survival in HGSC (HR, 0.4864; P = 0.0007). Multicolor immunohistochemistry and flow cytometry revealed a high degree of PD-1 and CD103 coexpression, specifically within the CD8 TIL compartment. PD-1(+)CD103(+) CD8 TIL were quiescent when assessed directly ex vivo yet were capable of robust cytokine production after pharmacologic stimulation. Moreover, they showed negligible expression of additional exhaustion-associated markers, including TIM-3, CTLA-4, and LAG-3. Thus, as hypothesized, CD103(+) CD8 TIL express PD-1 and appear quiescent in the tumor microenvironment. However, these cells retain functional competence and demonstrate strong prognostic significance. We speculate that, after standard treatment, PD-1(+)CD103(+) CD8 TIL might regain functional antitumor activity, an effect that potentially could be augmented by immune modulation.


Subject(s)
Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/immunology , Integrin alpha Chains/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Programmed Cell Death 1 Receptor/metabolism , Antigens, CD/genetics , CD8-Positive T-Lymphocytes/metabolism , Female , Gene Expression , Humans , Immunomodulation , Immunophenotyping , Integrin alpha Chains/genetics , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasm Grading , Neoplasm Staging , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Prognosis , Programmed Cell Death 1 Receptor/genetics , Retrospective Studies , Survival Analysis
13.
Cancer Immunol Res ; 3(3): 245-53, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25480168

ABSTRACT

CD25, the alpha subunit of the IL2 receptor, is a canonical marker of regulatory T cells (Treg) and hence has been implicated in immune suppression in cancer. However, CD25 is also required for optimal expansion and activity of effector T cells in peripheral tissues. Thus, we hypothesized that CD25, in addition to demarcating Tregs, might identify effector T cells in cancer. To investigate this possibility, we used multiparameter flow cytometry and IHC to analyze tumor-infiltrating lymphocytes (TIL) in primary high-grade serous carcinomas, the most common and fatal subtype of ovarian cancer. CD25 was expressed primarily by CD4⁺ TIL, with negligible expression by CD8⁺ TIL. In addition to conventional CD25⁺FoxP3⁺ Tregs, we identified a subset of CD25⁺FoxP3⁻ T cells that comprised up to 13% of CD4⁺ TIL. In tumors with CD8⁺ TIL, CD25⁺FoxP3⁻ T cells showed a strong positive association with patient survival (HR, 0.56; P = 0.02), which exceeded the negative effect of Tregs (HR, 1.55; P = 0.09). Among CD4⁺ TIL subsets, CD25⁺FoxP3⁻ cells expressed the highest levels of PD-1. Moreover, after in vitro stimulation, they failed to produce common T-helper cytokines (IFNγ, TNFα, IL2, IL4, IL10, or IL17A), suggesting that they were functionally exhausted. In contrast, the more abundant CD25⁻FoxP3⁻ subset of CD4⁺ TIL expressed low levels of PD-1 and produced T-helper 1 cytokines, yet conferred no prognostic benefit. Thus, CD25 identifies a subset of CD4⁺FoxP3⁻ TIL that, despite being exhausted at diagnosis, have a strong, positive association with patient survival and warrant consideration as effector T cells for immunotherapy.


Subject(s)
Forkhead Transcription Factors/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Ovarian Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Aged , Aged, 80 and over , CD8-Positive T-Lymphocytes/immunology , Cytokines/immunology , Female , Flow Cytometry , Humans , Middle Aged , Prognosis
14.
Oncoimmunology ; 3: e27668, 2014.
Article in English | MEDLINE | ID: mdl-25101220

ABSTRACT

Although high levels of tumor-infiltrating lymphocytes (TILs) generally correlate with good prognosis in high-grade serous ovarian cancer (HGSC) patients, little is known about the phenotype or specificity of these cells. We have recently demonstrated that TIL expressing the intra-epithelial lymphocyte marker CD103 (official name, integrin αE, ITGAE) abundantly infiltrate HGSCs, strongly correlating with increased disease-specific survival.

15.
Clin Cancer Res ; 20(5): 1125-34, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24323902

ABSTRACT

PURPOSE: Cancers accumulate mutations over time, each of which brings the potential for recognition by the immune system. We evaluated T-cell recognition of the tumor mutanome in patients with ovarian cancer undergoing standard treatment. EXPERIMENTAL DESIGN: Tumor-associated T cells from 3 patients with ovarian cancer were assessed by ELISPOT for recognition of nonsynonymous mutations identified by whole exome sequencing of autologous tumor. The relative levels of mutations and responding T cells were monitored in serial tumor samples collected at primary surgery and first and second recurrence. RESULTS: The vast majority of mutations (78/79) were not recognized by tumor-associated T cells; however, a highly specific CD8(+) T-cell response to the mutation hydroxysteroid dehydrogenase-like protein 1 (HSDL1)(L25V) was detected in one patient. In the primary tumor, the HSDL1(L25V) mutation had low prevalence and expression, and a corresponding T-cell response was undetectable. At first recurrence, there was a striking increase in the abundance of the mutation and corresponding MHC class I epitope, and this was accompanied by the emergence of the HSDL1(L25V)-specific CD8(+) T-cell response. At second recurrence, the HSDL1(L25V) mutation and epitope continued to be expressed; however, the corresponding T-cell response was no longer detectable. CONCLUSION: The immune system can respond to the evolving ovarian cancer genome. However, the T-cell response detected here was rare, was transient, and ultimately failed to prevent disease progression. These findings reveal the limitations of spontaneous tumor immunity in the setting of standard treatments and suggest a high degree of ignorance of tumor mutations that could potentially be reversed by immunotherapy.


Subject(s)
Immunologic Surveillance , Mutation , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Progression , Epitopes, T-Lymphocyte/immunology , Female , HLA Antigens/immunology , Humans , Hydroxysteroid Dehydrogenases/genetics , Immunohistochemistry , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasm Grading , Ovarian Neoplasms/pathology , Recurrence
16.
Clin Cancer Res ; 20(2): 434-44, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24190978

ABSTRACT

BACKGROUND: The presence of CD8(+) tumor-infiltrating lymphocytes (TIL) is associated with prolonged survival in high-grade serous ovarian cancer (HGSC) and other epithelial cancers. Survival is most strongly associated with intraepithelial versus intrastromal CD8(+) TILs; however, the mechanisms that promote the intraepithelial localization of TILs remain poorly understood. We hypothesized that intraepithelial CD8(+) TILs, like normal mucosal intraepithelial lymphocytes, might express CD103, a subunit of αE/ß7 integrin, which binds E-cadherin on epithelial cells. METHODS: A large collection of primary ovarian tumors (HGSC, endometrioid, mucinous, and clear cell) was analyzed by immunohistochemistry for the presence of TIL-expressing CD103. The activation and differentiation status of CD103(+) TILs were assessed by flow cytometry. The prognostic significance of TIL subsets was evaluated by Kaplan-Meier analysis. RESULTS: CD103(+) TILs were present in all major ovarian cancer subtypes and were most abundant in HGSC. CD103(+) TILs were preferentially localized to epithelial regions of tumors and were comprised predominantly of CD8(+) T cells expressing activation (HLA-DR, Ki-67, PD-1) and cytolytic (TIA-1) markers, as well as CD56(+) NK cells. Tumor infiltration by CD103(+) TILs was strongly associated with patient survival in HGSC. Tumors containing CD8(+) TILs that were CD103(-) showed poor prognosis equivalent to tumors lacking CD8(+) TILs altogether. CONCLUSIONS: CD103(+) TILs comprise intraepithelial, activated CD8(+) T cells, and NK cells and are strongly associated with patient survival in HGSC. CD103 may serve as a useful marker for enriching the most beneficial subsets of TILs for immunotherapy.


Subject(s)
Antigens, CD/metabolism , Cystadenocarcinoma, Serous/immunology , Cystadenocarcinoma, Serous/metabolism , Integrin alpha Chains/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , CD56 Antigen/immunology , CD56 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cystadenocarcinoma, Serous/mortality , Cystadenocarcinoma, Serous/pathology , Epithelium/immunology , Epithelium/metabolism , Female , Humans , Immunohistochemistry , Immunologic Memory , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Middle Aged , Neoplasm Grading , Neoplasm Staging , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology
17.
J Pathol ; 229(4): 515-24, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22996961

ABSTRACT

High-grade serous carcinoma (HGSC) is the most common and fatal form of ovarian cancer. While most tumours are highly sensitive to cytoreductive surgery and platinum- and taxane-based chemotherapy, the majority of patients experience recurrence of treatment-resistant tumours. The clonal origin and mutational adaptations associated with recurrent disease are poorly understood. We performed whole exome sequencing on tumour cells harvested from ascites at three time points (primary, first recurrence, and second recurrence) for three HGSC patients receiving standard treatment. Somatic point mutations and small insertions and deletions were identified by comparison to constitutional DNA. The clonal structure and evolution of tumours were inferred from patterns of mutant allele frequencies. TP53 mutations were predominant in all patients at all time points, consistent with the known founder role of this gene. Tumours from all three patients also harboured mutations associated with cell cycle checkpoint function and Golgi vesicle trafficking. There was convergence of germline and somatic variants within the DNA repair, ECM, cell cycle control, and Golgi vesicle pathways. The vast majority of somatic variants found in recurrent tumours were present in primary tumours. Our findings highlight both known and novel pathways that are commonly mutated in HGSC. Moreover, they provide the first evidence at single nucleotide resolution that recurrent HGSC arises from multiple clones present in the primary tumour with negligible accumulation of new mutations during standard treatment.


Subject(s)
Clonal Evolution/genetics , Cystadenocarcinoma, Serous/genetics , Neoplasm Recurrence, Local/genetics , Ovarian Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Alleles , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/pathology , DNA, Neoplasm/genetics , Drug Resistance, Neoplasm , Exome , Extracellular Matrix/genetics , Female , Gene Regulatory Networks , Genomics , Golgi Apparatus/genetics , Humans , Mutation , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Sequence Analysis, DNA
18.
PLoS One ; 7(3): e32805, 2012.
Article in English | MEDLINE | ID: mdl-22431983

ABSTRACT

Nitrotyrosination of proteins, a hallmark of inflammation, may result in the production of MHC-restricted neoantigens that can be recognized by T cells and bypass the constraints of immunological self-tolerance. Here we biochemically and structurally assessed how nitrotyrosination of the lymphocytic choriomeningitis virus (LCMV)-associated immunodominant MHC class I-restricted epitopes gp33 and gp34 alters T cell recognition in the context of both H-2D(b) and H-2K(b). Comparative analysis of the crystal structures of H-2K(b)/gp34 and H-2K(b)/NY-gp34 demonstrated that nitrotyrosination of p3Y in gp34 abrogates a hydrogen bond interaction formed with the H-2K(b) residue E152. As a consequence the conformation of the TCR-interacting E152 was profoundly altered in H-2K(b)/NY-gp34 when compared to H-2K(b)/gp34, thereby modifying the surface of the nitrotyrosinated MHC complex. Furthermore, nitrotyrosination of gp34 resulted in structural over-packing, straining the overall conformation and considerably reducing the stability of the H-2K(b)/NY-gp34 MHC complex when compared to H-2K(b)/gp34. Our structural analysis also indicates that nitrotyrosination of the main TCR-interacting residue p4Y in gp33 abrogates recognition of H-2D(b)/gp33-NY complexes by H-2D(b)/gp33-specific T cells through sterical hindrance. In conclusion, this study provides the first structural and biochemical evidence for how MHC class I-restricted nitrotyrosinated neoantigens may enable viral escape and break immune tolerance.


Subject(s)
Inflammation/immunology , Major Histocompatibility Complex/immunology , Receptors, Antigen, T-Cell/immunology , Tyrosine/analogs & derivatives , Amino Acids/metabolism , Animals , Crystallography, X-Ray , H-2 Antigens/chemistry , H-2 Antigens/immunology , Lymphocytic choriomeningitis virus/immunology , Mice , Models, Molecular , Peptides/chemistry , Peptides/metabolism , Protein Conformation , Protein Stability , Receptors, Antigen, T-Cell/chemistry , Tyrosine/immunology , Viral Proteins/chemistry , Viral Proteins/immunology
19.
J Transl Med ; 10: 33, 2012 Feb 27.
Article in English | MEDLINE | ID: mdl-22369276

ABSTRACT

BACKGROUND: The immune system strongly influences outcome in patients with ovarian cancer. In particular, the absolute lymphocyte count in peripheral blood (ALC) and the presence of tumor-infiltrating lymphocytes (TIL) have each been associated with favourable prognosis. However, the mechanistic relationships between ALC, TIL and prognosis are poorly understood. We hypothesized that high ALC values might be associated with stronger tumor immunity as manifested by increased TIL, decreased tumor burden and longer survival. METHODS: ALC values were collected from patient records ≥ 2 years before, during or after primary treatment for high-grade serous ovarian cancer (HGSC). Lymphocyte subsets were assessed in peripheral blood by flow cytometry. CD8+ and CD20+ TIL were assessed by immunohistochemistry. RESULTS: Overall, patients had normal ALC values two or more years prior to diagnosis of HGSC. These values were not predictive of disease severity or survival upon subsequent development of HGSC. Rather, ALC declined upon development of HGSC in proportion to disease burden. This decline involved all lymphocyte subsets. ALC increased following surgery, remained stable during chemotherapy, but rarely recovered to pre-diagnostic levels. ALC values recorded at diagnosis did not correlate with CD8+ or CD20+ TIL but were associated with progression-free survival. CONCLUSIONS: Patients with high intrinsic ALC values show no clinical or survival advantage upon subsequent development of HGSC. ALC values at diagnosis are prognostic due to an association with disease burden rather than TIL. Therapeutic enhancement of ALC may be necessary but not sufficient to improve survival in HGSC.


Subject(s)
Lymphocytes, Tumor-Infiltrating/immunology , Ovarian Neoplasms/immunology , Cohort Studies , Cost of Illness , Female , Flow Cytometry , Humans , Lymphocyte Count , Lymphocyte Subsets/immunology , Middle Aged , Neoplasm Grading , Neoplasms, Cystic, Mucinous, and Serous/drug therapy , Neoplasms, Cystic, Mucinous, and Serous/immunology , Neoplasms, Cystic, Mucinous, and Serous/pathology , Neoplasms, Cystic, Mucinous, and Serous/surgery , Neutrophils/immunology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/surgery , Prognosis , Survival Analysis , Treatment Outcome
20.
Vaccine ; 29(44): 7857-66, 2011 Oct 13.
Article in English | MEDLINE | ID: mdl-21816200

ABSTRACT

Persistent infection by high risk genotypes of human papillomavirus (HPV) is the cause of cervical cancer, which remains one of the most common cancers among women worldwide. In addition, there is a growing appreciation that high risk HPVs are associated with a number of other cancers including anogenital cancers as well as a subset of head and neck cancers. Recently, prophylactic HPV vaccines targeting the two most prevalent high risk HPVs (HPV16 and HPV18) have been deployed in large-scale vaccination campaigns. However, the extent to which these prophylactic vaccines confer protection against other high risk HPV genotypes is largely unknown and prophylactic vaccines have been shown to be ineffective against pre-existing infection. Thus there continues to be an urgent need for effective therapeutic vaccines against HPV. The E7 protein of HPV16 has been widely studied as a target for therapeutic vaccines in HPV-associated cancer settings because HPV16 is the most prevalent of the high risk HPV genotypes. However, HPV16 accounts for only about 50% of cervical cancers and there are at least 15 other high risk HPVs that are known to be oncogenic. We have developed a novel, broad-spectrum, therapeutic vaccine (Pentarix) directed at the E7 proteins from five of the most prevalent high-risk genotypes of HPV worldwide (HPV16, 18, 31, 45 and 52) that together account for more than 80% of all HPV-associated cancers. Pentarix is a recombinant protein-based vaccine that elicits strong, multi-genotype specific CD8 T cell immunity when administered to mice in combination with adjuvants comprised of agonists of the TLR3 or TLR9 family of innate immune receptors. Furthermore, large, established E7-expressing TC-1 tumors undergo rapid and complete regression after therapeutic vaccination of mice with Pentarix. Together, these data suggest that Pentarix may be of clinical value for patients with E7-positive, HPV-associated precancerous lesions or malignant disease.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carcinoma/therapy , Immunotherapy/methods , Papillomavirus E7 Proteins/immunology , Papillomavirus Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Amino Acid Sequence , Animals , Carcinoma/mortality , Carcinoma/pathology , Female , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Papillomavirus Vaccines/administration & dosage , Sequence Alignment , Survival Analysis , Treatment Outcome , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...