GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Cancers, MDPI AG, Vol. 14, No. 13 ( 2022-06-28), p. 3159-
    Abstract: Cyclin-dependent kinase 4/6 inhibitors are the standard of care for hormone receptor-positive metastatic breast cancer. This retrospective study reports on genomic biomarkers of CDK 4/6i resistance utilizing genomic data acquired through routine clinical practice. Patients with HR+ MBC treated with palbociclib, ribociclib, or abemaciclib and antiestrogen therapy were identified. Patients were grouped into early ( 〈 6 months); intermediate (6–24 months for 0–1 lines; 6–9 months for ≥2 lines); or late progressors ( 〉 24 months for 0–1 lines; 〉 9 months PFS for ≥2 lines). NGS and RNA sequencing data were analyzed in association with PFS, and survival analysis was stratified by prior lines of chemotherapy. A total of 795 patients with HR+ MBC treated with CDK 4/6i were identified. Of these, 144 (18%) patients had genomic data and 29 (3.6%) had RNA data. Among the 109 patients who received CDK4/6i as 1st- or 2nd-line therapy, 17 genes showed associations with PFS (p-value ≤ 0.15 and HR ≥ 1.5 or HR 〈 0.5). Whole transcriptome RNAseq was analyzed for 24/109 (22%) patients with 0–1 prior lines of therapy and 56 genes associated with PFS (HR ≥ 4 or HR ≤ 0.25 and FDR ≤ 0.15). In this retrospective analysis, genomic biomarkers including FGFR1 amplification, PTEN loss, and DNA repair pathway gene mutations showed significant associations with shorter PFS for patients receiving CDK4/6 inhibitor therapy.
    Type of Medium: Online Resource
    ISSN: 2072-6694
    Language: English
    Publisher: MDPI AG
    Publication Date: 2022
    detail.hit.zdb_id: 2527080-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2017
    In:  The Journal of Immunology Vol. 198, No. 1_Supplement ( 2017-05-01), p. 196.11-196.11
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 198, No. 1_Supplement ( 2017-05-01), p. 196.11-196.11
    Abstract: CD8+ Tumor Infiltrating Lymphocytes (TILs) have been shown to correlate with patient prognosis in breast cancer. Recently, resident memory T cells (TRM) have been established as a potent functional subset of memory T cells that exist in peripheral tissue without recirculation. We assayed CD8+ T cells from fresh breast tumors to phenotype memory T cells and found them to be almost exclusively made up of effector memory T cells. Further profiling by examining expression of CD103 and CD69 showed that on average 40% of CD8+ TILs are composed of TRM. Functional analysis of these T cells showed robust production of IFNγ and TNFα by these TRMs suggesting that despite existence in the tumor microenvironment TRMs maintain functional potency. To examine if CD8+ TRM have prognostic value in breast cancer patients, we examined a set of primary tumors from ‘good’ and ‘bad’ outcome patients, which we defined as having a relapse in more than 5 years after diagnosis or in less than 3 years after diagnosis respectively. In good outcome patients TRMs represented 60% of CD8+ T cells found in cancer islands, while only 20% of such in bad outcome patients. Similarly, CD8+ TRMs were observed in higher numbers in both the cancer islands and stroma of good outcome patient tumors in comparison to bad outcome patient tumors. This observed significance was greater than that found for CD8+ T cells in general, highlighting the importance of TRM in affecting patient outcome. Finally we showed that TRMs also exist in non-cancerous breast tissue and are found within the mammary ducts associated with epithelial cells perhaps in surveillance of potential pathogens. We suggest that TRMs are a potential vaccine target for preventing recurrence and metastasis in cancer patients.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2017
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2018
    In:  The Journal of Immunology Vol. 200, No. 1_Supplement ( 2018-05-01), p. 57.23-57.23
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 200, No. 1_Supplement ( 2018-05-01), p. 57.23-57.23
    Abstract: CD8+ T cell infiltration into solid tumors has been shown to correlate with patient prognosis in numerous carcinomas. Furthermore, response to checkpoint blockade therapies has been linked to an induction of T cell function and rescue of T cell exhaustion in patients. Expression of both PD-1 and CD39 on exhausted CD8+ T cells has been described in murine models of chronic disease and recently human carcinomas. Here we profile breast tumor, melanoma, and chronic lymphocytic leukemia (CLL) for the frequency and phenotype of exhausted CD8+ T cells. We find that the frequency of exhausted CD8+ T cells is highest in melanoma among these tumor types which may suggest correlation with mutational burden. Functional analysis of PD-1+ CD39+ cells in human tumors shows significant reduction of IFNγ, TNFα, and IL-2 production as assessed by intracellular flow cytometry. Further phenotyping of exhausted CD8+ T cells revealed a loss of expression of both the IL-7 receptor alpha (CD127) and KLRG1, suggesting terminal differentiation of these cells. PD-1+ CD39+ T cells also expressed CD103 suggesting potential similarities with differentiation into a peripheral resident memory phenotype. Indeed PD-1+ CD39+ exhausted T cells were only identified in peripheral tissues where cancer cells were present, such as solid tumors, tumor positive tumor draining lymph nodes, and one CLL patient, but not identified in the circulation or tumor negative tumor draining lymph nodes of patients. Finally, single cell and T cell receptor sequencing revealed distinct transcriptional and clonal signatures of PD-1+ CD39+ CD8+ T cells suggesting them to be a unique population in response to tumor antigen.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2018
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 6788-6788
    Abstract: Background: CF33-hNIS-anti-PD-L1 is a novel chimeric orthopoxvirus shown to have anti-cancer activity in triple negative breast cancer (TNBC) xenografts. For clinical tracking of the oncolytic virus (OV), human sodium iodide symporter (hNIS) transgene was inserted into the virus. hNIS gene expression allows cells to take up iodine and be visible by non-invasive imaging techniques. Animal studies showed that tumor cells infected with CF33-hNIS-anti-PD-L1 express functional hNIS and are visible by single-photon emission computed tomography (SPECT) or positron emission tomography (PET). The current report describes imaging results from an ongoing first-in-human trial. Methods: This is a phase I, single center, single arm clinical trial evaluating the safety and tolerability of CF33-hNIS-anti-PD-L1 intratumoral (IT) injections in patients with metastatic TNBC. Key eligibility criteria include patients with unresectable or metastatic TNBC; progressed on at least 2 prior chemotherapies; ECOG 0-2; RECIST 1.1 measurable disease; and at least one tumor amenable to repeated IT injections. Eligible patients receive CF33-hNIS-anti-PD-L1 IT at 1 of 8 assigned dose levels (from 1 × 105 PFU to 3 x 108 PFU) on Days 1 and 15 of each 28-day cycle for a total of 3 cycles of treatment. The primary objective is to evaluate the safety and tolerability of CF33-hNIS-anti-PD-L1. Secondary objectives are to determine optimal biological dose, recommended phase II dose, and response rates. Exploratory objectives include assessing feasibility of non-invasive hNIS imaging as a method of tracking viral infection and replication. SPECT whole body imaging was performed at Cycle 1 Day 8 (C1D8). Results: From October 2021 to October 2022, 8 patients were enrolled in this ongoing study and received at least 1 dose of CF33-hNIS-anti-PD-L1 injection at one of the first 3 dose levels (1 x 105, 3 x 105, or 1 x 106 PFU). All 8 patients underwent SPECT imaging at C1D8 with 6/8 patients (75%) having uptake at the site of injection on the SPECT imaging study. Four of 4 patients (100%) with injection sites at metastatic subcutaneous nodules, intramuscular masses, or axillary lymph nodes and 2/4 patients (50%) with injection sites at matted dermal metastatic lesions had uptake on SPECT. Conclusion: SPECT imaging successfully showed enhancement at the injected lesions in 75% of patients treated with CF33-hNIS-anti-PD-L1, even at current low doses, suggesting local viral replication and hNIS expression. Further analysis will evaluate the correlation of SPECT imaging results with pathologic immune cell infiltrate, viral staining, and tumor response. This is the first known report of hNIS-based imaging to track oncolytic poxvirus replication in humans and gives promise that this technology may be used for noninvasive tracking of systemically administered OV and other therapies. Citation Format: Jamie Green Rand, Dave Yamauchi, Shyambabu Chaurasiya, Jianying Zhang, Raju Pillai, Colt Egelston, Jonathan Kessler, Badri Modi, Leslie Chong, Amanda Seiz, Giovanni Selvaggi, Nick Ede, Mireya Murga, Norma Martinez, Wichanee Borisuthirattana, Hans Meisen, Susan Yost, James Waisman, Daphne Stewart, Joanne Mortimer, Yuman Fong, Yuan Yuan. hNIS imaging data from a first-in-human trial of the oncolytic virus CF33-hNIS-antiPD-L1 in patients with triple negative breast cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6788.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2021
    In:  Cancer Research Vol. 81, No. 4_Supplement ( 2021-02-15), p. PS19-16-PS19-16
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 4_Supplement ( 2021-02-15), p. PS19-16-PS19-16
    Abstract: Metastatic disease is understudied largely because of inaccessibility to quality specimens for research use. The Legacy Project, a rapid or “warm”, autopsy program at City of Hope, seeks to overcome this challenge by collecting tissue from metastatic patients immediately (within 6 hours) after their death. This paradigm serves as a specimen resource to address many clinically relevant questions, such as disease heterogeneity and mechanisms driving disease progression. Using this model, we uncovered clinically relevant disease information that is normally unavailable while a subject is alive. In this study, 9 metastatic breast cancer patients and their families were approached and consented prior to death. The cohort includes a diversity of clinical presentations in terms of disease subtype, progression history, organ involvement, and final cause of death. A total of 533 specimens were collected across 9 subjects. The average time from death to specimen acquisition was 6.1 hours (range: 4.03 - 7.66 hours; median: 5.71 hours). Total number of specimens collected from each participant ranged from 38-75, with an average of 60 across all patients; the mean number of tumor-positive specimens collected was 29 (range 12-46); the mean number of non-cancer specimens collected was 31 (range 25-45). In patients with primary estrogen receptor (ER) positive disease, we observed variable heterogeneity in estrogen, progesterone, and ki67 status across metastatic lesions. Furthermore, we observed a profound shift in disease phenotype towards end of life, trending towards complete loss of hormone receptor expression and stark increase of Ki67 levels. At the time of procurement, one third of subjects exhibited clinically unidentified diseased sites in organs not commonly associated with breast cancer metastases a, including ovary, kidney, and pancreas. In two other instances, “resolved” bone specimens (as measured by absence of FTG uptake in PET/CT imaging) were later determined to be & gt;30% tumor positive when assessed by H & E. While these preliminary findings generate more questions than answers regarding mechanisms of metastatic progression and resistance to therapy, they highlight the utility of rapid autopsy in a research setting. We suggest that many unanswered clinical questions can be addressed through interrogation of post-mortem tissues and we urge research institutions to thoughtfully consider adoption of the “rapid autopsy” model. Citation Format: Eliza R Bacon, Kena Ihle, Colt Egelston, Weihua Guo, Diana Simons, Peter P Lee, James Waisman. Utility of rapid autopsy in cancer research: Unexpected findings and lessons learned from warm autopsies of metastatic breast cancer patients [abstract]. In: Proceedings of the 2020 San Antonio Breast Cancer Virtual Symposium; 2020 Dec 8-11; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2021;81(4 Suppl):Abstract nr PS19-16.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 5_Supplement ( 2023-03-01), p. OT2-08-03-OT2-08-03
    Abstract: Background: Combination of immune checkpoint inhibitor pembrolizumab and chemotherapy are standard of care therapy for patients with programmed death-ligand 1 positive (PD-L1+) metastatic triple negative breast cancer (mTNBC). However, a greater understanding of how immune checkpoint inhibitors and chemotherapies synergize to yield anti-tumor T cell responses is needed. The current phase I study evaluated the immunogenicity of doxorubicin plus pembrolizumab in patients with mTNBC. Patients and Methods: Patients with mTNBC, no prior anthracycline use, and 0-2 lines of prior systemic chemotherapies received pembrolizumab 200 mg IV and doxorubicin 50-60 mg/m2 IV every 3 weeks for 6 cycles followed by pembrolizumab maintenance until disease progression or intolerance. Patients were not selected based on PD-L1 expression. The primary objectives were safety and objective response rate per RECIST 1.1. Peripheral blood samples were collected at baseline, Cycle 2 Day 1 (C2D1), and post Cycle 3 for analysis by high parameter flow cytometry. Results: Ten patients were enrolled between March 2016 and November 2019. Best responses included one patient with complete response (CR), five with partial responses (PR), two with stable disease (SD), and one with progression of disease (PD). Flow cytometry showed increased CD3+ T cells (p=0.03) from pre-treatment to C2D1 with substantial inter-patient variation in CD3+ T cells. The expansion of proliferative exhausted-like PD-1+ CD8+ T cell population was identified in 8/9 patients, and exhausted CD8+ T cells were significantly expanded from pre-treatment to C2D1 in the patient with CR (p=0.01). Notably, frequencies of PD-1 high proliferative CD8+ T cells contracted significantly from C2D1 to post Cycle 3 (p=0.005), with a return to near baseline frequencies in the majority of the patients. In the context of all identified T cell subsets, PD-1hi proliferative CD8+ T cells demonstrated the greatest increase in fold change from pre-treatment to C2D1. In contrast, PD-1lo proliferative CD8+ T cells demonstrated the greatest decrease in fold change from pre-treatment to C3D1. Conclusion: Anthracycline-naïve patients with mTNBC treated with the combination of pembrolizumab and doxorubicin yielded robust peripheral blood T cell responses. Further studies dissecting the dynamics and durability of anti-tumor T cell responses and concurrent tumor immune microenvironment changes are needed to optimize combined immune checkpoint inhibitor and chemotherapy treatment for patients with mTNBC. Citation Format: Colt A. Egelston, Weihua Guo, Susan E. Yost, Xuan Ge, Jin Sun Lee, Paul H. Frankel, Yujie Cui, Christopher Ruel, Daniel Schmolze, Mireya Murga, Aileen Tang, Norma Martinez, Misagh Karimi, George Somlo, Peter Lee, James Waisman, Yuan Yuan. Immunogenicity of pembrolizumab and doxorubicin in a phase I trial for patients with metastatic triple negative breast cancer [abstract]. In: Proceedings of the 2022 San Antonio Breast Cancer Symposium; 2022 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2023;83(5 Suppl):Abstract nr OT2-08-03.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2014
    In:  The Journal of Immunology Vol. 192, No. 1_Supplement ( 2014-05-01), p. 71.46-71.46
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 192, No. 1_Supplement ( 2014-05-01), p. 71.46-71.46
    Abstract: Tumor tissues are composed of significant numbers of Tumor Infiltrating Lymphocytes (TILs), which may include CD8+ T cells, CD4+ T cells, and B cells. Of the T cell population, it is known that these cells are often functionally suppressed and defective in their ability to kill and proliferate. However, little is known about how T cells traffic into tumor sites. Furthermore, it is unclear how the various T cell memory subtypes play a role in attempting to eradicate tumor cells. Such knowledge is critical for the development of successful cancer immunotherapies. Here we assayed peripheral blood mononuclear cells (PBMCs), tumor tissue, and sentinel lymph nodes (SLNs) of breast cancer patients to phenotype memory T cells. We found higher proportions of EM cells among CD8 TILs as compared to SLN CD8 T cells. Conversely, the CD4 compartment in both TILs and SLNs was dominated by CM CD4 T cells. Further analysis showed that breast cancer patient CD8+ TILs in primary tumors were made up of a significant population of Tissue Resident Memory cells (TRMs) as identified by CD103 expression and high CD69 expression. We also showed that the CD8+ TILs found in a metastatic brain lesion of a breast cancer patient also contained TRMs. Finally we showed that healthy breast tissue from a patient with no clinical signs of breast cancer also contained CD8+ TRMs.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2014
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2009
    In:  The Journal of Immunology Vol. 182, No. 1_Supplement ( 2009-04-01), p. 95.5-95.5
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 182, No. 1_Supplement ( 2009-04-01), p. 95.5-95.5
    Abstract: Rheumatoid arthritis (RA) is largely mediated by autoantibodies, the production of which is supported by autoreactive T cells. Previous reports in RA and its animal models also suggest that T cells present in the joints can locally propagate arthritis. To identify T cells migrating to the joints before and during arthritis development, we transferred fluorescence-labeled T cells and unlabeled antigen-presenting cells from BALB/c mice with proteoglycan-induced arthritis (PGIA) to syngeneic immunodeficient mice, and then monitored the recruitment of donor T cells in the ankle joints of the recipients using in vivo two-photon microscopy, immunohistochemistry, and flow cytometry. Despite the development of arthritis in the recipient mice, we found very few donor T cells in their joints after cell transfer. Furthermore, treatment of recipient mice with FTY720, an inhibitor of lymphocyte egress from lymphoid organs, which effectively depleted T cells in both peripheral blood and joints, failed to inhibit PGIA development. In contrast, arthritis did not occur in recipient mice after injection with donor cells depleted in T cells in vitro prior to transfer. Our results suggest that while lymphoid organ-homing T cells are required for the adoptive transfer of PGIA, the few T cells that migrate to the joints do not play a significant role in arthritis development. Work supported by the NIH and the Grainger Foundation.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2009
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2011
    In:  The Journal of Immunology Vol. 186, No. 1_Supplement ( 2011-04-01), p. 167.21-167.21
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 186, No. 1_Supplement ( 2011-04-01), p. 167.21-167.21
    Abstract: Joint damage in rheumatoid arthritis (RA) is largely mediated by synovial fluid (SF) neutrophils. In proteoglycan (PG)-induced arthritis (PGIA), an autoimmune mouse model of RA, SF cells show high expression of the myeloid lineage markers CD11b and Gr-1. The effects of this neutrophil-like SF cell population on the activation of dendritic cells (DCs) or T cells have not been studied in RA or its animal models. We co-cultured SF cells of PGIA mice with bone marrow-derived DCs, and found that levels of MHC II and CD86 expression by DCs decreased significantly in the presences of SF cells. To determine if SF cells also reduced the antigen (Ag)-presenting capacity of the DCs, we added naïve PG-specific TCR transgenic T cells and cognate Ag to the co-cultures. T cells cultured in the presence of SF cells and Ag-loaded DCs showed significant decreases in CD69 expression and proliferation compared to T cells cultured with Ag-loaded DCs only. Arginase-1, a marker of myeloid-derived suppressor cells (MDSCs) was upregulated at both transcriptional and protein levels in SF cells. Expression and activity of arginase-1 were also elevated in spleen cells of arthritic mice as compared to spleen cells of naïve non-arthritic mice. Our results suggest that SF cells in the arthritic joints and spleens of mice with PGIA contain a population of MDSCs, which might have the capacity to suppress autoreactive T cell responses and thus accelerate the resolution of joint inflammation.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2011
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2019
    In:  The Journal of Immunology Vol. 202, No. 1_Supplement ( 2019-05-01), p. 138.21-138.21
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 202, No. 1_Supplement ( 2019-05-01), p. 138.21-138.21
    Abstract: Tumor immunogenicity is generally thought to be correlated with increased tumor mutation burden and the resulting neoantigen driven T cell reactivity to cancer cells. This immunogenicity results in the formation of tumor specific, but exhausted tumor infiltrating CD8+ T cells (TILex). Breast cancer tumors, which are predominantly low mutation burden, are widely viewed as non-immunogenic. Here we demonstrate that subsets of both triple negative and estrogen receptor positive breast cancer patients have primary tumors heavily infiltrated by tumor specific CD8+ TILex. CD8+ TILex in breast cancer patients were demonstrated to coexpress PD-1 and CD39 and showed a significant reduction of IFNγ, TNFα, and IL-2 production capacity. Like melanoma and colorectal CD8+ TILex, CD8+ TILex human breast tumors had increased expression of CD103, CD38, and TIM-3 in addition to loss of KLRG1 and CD127 expression. Similarly, melanoma, colorectal, and breast tumor CD8+ TILex shared transcriptional signatures identified by single cell sequencing. T cell receptor sequencing revealed a distinct repertoire of CD8+ TILex as compared to other CD8+ TILs from the same tumors, suggesting them to be a unique population in response to tumor antigen. Finally, we demonstrated that increased levels of CD8+ TILex in human breast tumors are not associated with higher levels of tumor mutation burden, as assessed by a tumor mutation load oncogene panel. This work suggests that a subset of breast cancer patients have inflamed, tumors associated with the presence of exhausted CD8+ T cells and that these patients may benefit from immunotherapeutic interventions.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2019
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...