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: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 1068-1068
    Abstract: Introduction: Adoptive transfer of tumor infiltrating lymphocytes (TIL) can mediate long-term durable regression in patients with metastatic melanoma, a type of cancer which is characterized by a high number of mutated genes and pronounced lymphocytic infiltrate. In contrast, metastatic breast cancer expresses far fewer somatic mutations and may contain lower levels of TIL. This pilot study investigated the ability to identify immunogenic non-synonymous somatic mutations which can serve as targets for adoptively transferred autologous T cells into patients with metastatic breast cancer, refractory to other treatments. Methods: WES and RNAseq were performed on metastatic lesions resected in the Surgery branch, NCI, NIH from patients with metastatic breast cancer. Non-synonymous somatic mutations were identified and, in parallel, TIL from the same lesions were grown ex vivo in the presence of IL-2. The somatic mutations were tested for recognition by the autologous TIL using previously described functional assays, involving tandem mini-genes (TMG), long peptides or predicted minimal peptide approaches. For some patients, the mutation-reactive T cells were sorted by FACS and the respective TCR was identified by single cell sequencing. Results: Metastatic tumor lesions from 37 patients with breast cancer were studied and all of them were found to contain and express non-synonymous somatic mutations (range: 4-1788, median: 102). TIL were successfully grown ex vivo from the tumors of all patients. Following functional assays (IFN-γ secretion and 4-1BB/OX-40 upregulation), it was found that autologous TIL recognized at least one (range: 1-11, median: 3) mutated product in 25 of 37 patients (68%) tested. Seventy-five percent of those were recognized by CD4+ and 25% by CD8+ T cells. All identified immunogenic tumor mutations (n=88) were unique and not shared by other patients. Sixty-four unique TCR pairs were isolated from the reactive cells and verified to recognize 30 immunogenic tumor mutations (range 1-7 TCRs/neoantigen). Conclusions: Patients with metastatic breast cancer contain a substantial number of non-synonymous somatic mutations and mutation-reactive TIL were found in the majority of those patients. The immunogenicity of tumor-specific mutations in metastatic breast cancer can be the platform for a highly personalized adoptive T cell transfer of selected TIL or mutation-reactive TCR transduced T cells targeting those tumor mutated genes. Citation Format: Nikolaos Zacharakis, Peter Kim, Paul Robbins, Jared Gartner, Todd Prickett, Victoria Hill, Biman Paria, Steven Feldman, Stephanie Goff, Steven Rosenberg. Tumor infiltrating lymphocytes (TIL) recognize a unique and diverse number of non-synonymous somatic mutations in patients with metastatic breast cancer [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 1068.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    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 ...
  • 2
    In: Journal of Clinical Investigation, American Society for Clinical Investigation, Vol. 129, No. 3 ( 2019-2-4), p. 1109-1114
    Type of Medium: Online Resource
    ISSN: 0021-9738 , 1558-8238
    Language: English
    Publisher: American Society for Clinical Investigation
    Publication Date: 2019
    detail.hit.zdb_id: 2018375-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 9, No. 7 ( 2021-07), p. e002882-
    Abstract: The adoptive transfer of naturally occurring T cells that recognize cancer neoantigens has led to durable tumor regressions in select patients with cancer. However, it remains unknown whether such T cells can be isolated from and used to treat patients with glioblastoma, a cancer that is refractory to currently available therapies. To answer this question, we stimulated patient blood-derived memory T cells in vitro using peptides and minigenes that represented point mutations unique to patients’ tumors (ie, candidate neoantigens) and then tested their ability to specifically recognize these mutations. In a cohort of five patients with glioblastoma, we found that circulating CD4 + memory T cells from one patient recognized a cancer neoantigen harboring a mutation in the EED gene (EED H189N ) that was unique to that patient’s tumor. This finding suggests that neoantigen-reactive T cells could indeed be isolated from patients with glioblastoma, thereby providing a rationale for further efforts to develop neoantigen-directed adoptive T cell therapy for this disease.
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2021
    detail.hit.zdb_id: 2719863-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Science, American Association for the Advancement of Science (AAAS), Vol. 370, No. 6522 ( 2020-12-11), p. 1328-1334
    Abstract: Adoptive T cell therapy (ACT) using ex vivo–expanded autologous tumor-infiltrating lymphocytes (TILs) can mediate complete regression of certain human cancers. The impact of TIL phenotypes on clinical success of TIL-ACT is currently unclear. Using high-dimensional analysis of human ACT products, we identified a memory-progenitor CD39-negative stem-like phenotype (CD39 − CD69 − ) associated with complete cancer regression and TIL persistence and a terminally differentiated CD39-positive state (CD39 + CD69 + ) associated with poor TIL persistence. Most antitumor neoantigen-reactive TILs were found in the differentiated CD39 + state. However, ACT responders retained a pool of CD39 − stem-like neoantigen-specific TILs that was lacking in ACT nonresponders. Tumor-reactive stem-like TILs were capable of self-renewal, expansion, persistence, and superior antitumor response in vivo. These data suggest that TIL subsets mediating ACT response are distinct from TIL subsets enriched for antitumor reactivity.
    Type of Medium: Online Resource
    ISSN: 0036-8075 , 1095-9203
    RVK:
    RVK:
    Language: English
    Publisher: American Association for the Advancement of Science (AAAS)
    Publication Date: 2020
    detail.hit.zdb_id: 128410-1
    detail.hit.zdb_id: 2066996-3
    detail.hit.zdb_id: 2060783-0
    SSG: 11
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 10, No. 8 ( 2022-08-03), p. 932-946
    Abstract: Adoptive cellular therapy (ACT) targeting neoantigens can achieve durable clinical responses in patients with cancer. Most neoantigens arise from patient-specific mutations, requiring highly individualized treatments. To broaden the applicability of ACT targeting neoantigens, we focused on TP53 mutations commonly shared across different cancer types. We performed whole-exome sequencing on 163 patients with metastatic solid cancers, identified 78 who had TP53 missense mutations, and through immunologic screening, identified 21 unique T-cell reactivities. Here, we report a library of 39 T-cell receptors (TCR) targeting TP53 mutations shared among 7.3% of patients with solid tumors. These TCRs recognized tumor cells in a TP53 mutation- and human leucocyte antigen (HLA)-specific manner in vitro and in vivo. Twelve patients with chemorefractory epithelial cancers were treated with ex vivo–expanded autologous tumor-infiltrating lymphocytes (TIL) that were naturally reactive against TP53 mutations. However, limited clinical responses (2 partial responses among 12 patients) were seen. These infusions contained low frequencies of mutant p53–reactive TILs that had exhausted phenotypes and showed poor persistence. We also treated one patient who had chemorefractory breast cancer with ACT comprising autologous peripheral blood lymphocytes transduced with an allogeneic HLA-A*02–restricted TCR specific for p53R175H. The infused cells exhibited an improved immunophenotype and prolonged persistence compared with TIL ACT and the patient experienced an objective tumor regression (-55%) that lasted 6 months. Collectively, these proof-of-concept data suggest that the library of TCRs targeting shared p53 neoantigens should be further evaluated for the treatment of patients with advanced human cancers. See related Spotlight by Klebanoff, p. 919
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 11, No. 5 ( 2023-05), p. e006264-
    Abstract: Cellular immunotherapies using autologous tumor-infiltrating lymphocytes (TIL) can induce durable regression of epithelial cancers in selected patients with treatment-refractory metastatic disease. As the genetic engineering of T cells with tumor-reactive T-cell receptors (TCRs) comes to the forefront of clinical investigation, the rapid, scalable, and cost-effective detection of patient-specific neoantigen-reactive TIL remains a top priority. Methods We analyzed the single-cell transcriptomic states of 31 neoantigen-specific T-cell clonotypes to identify cell surface dysfunction markers that best identified the metastatic transcriptional states enriched with antitumor TIL. We developed an efficient method to capture neoantigen-reactive TCRs directly from resected human tumors based on cell surface co-expression of CD39, programmed cell death protein-1, and TIGIT dysfunction markers (CD8 + TIL TP ). Results TIL TP TCR isolation achieved a high degree of correlation with single-cell transcriptomic signatures that identify neoantigen-reactive TCRs, making it a cost-effective strategy using widely available resources. Reconstruction of additional TIL TP TCRs from tumors identified known and novel antitumor TCRs, showing that at least 39.5% of TIL TP TCRs are neoantigen-reactive or tumor-reactive. Despite their substantial enrichment for neoantigen-reactive TCR clonotypes, clonal dynamics of 24 unique antitumor TIL TP clonotypes from four patients indicated that most in vitro expanded TIL TP populations failed to demonstrate neoantigen reactivity, either by loss of neoantigen-reactive clones during TIL expansion, or through functional impairment during cognate neoantigen recognition. Conclusions While direct usage of in vitro-expanded CD8 + TIL TP as a source for cellular therapy might be precluded by profound TIL dysfunction, isolating TIL TP represents a streamlined effective approach to rapidly identify neoantigen-reactive TCRs to design engineered cellular immunotherapies against cancer.
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2023
    detail.hit.zdb_id: 2719863-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 9, No. Suppl 2 ( 2021-11), p. A177-A177
    Abstract: As cellular immunotherapies utilizing genetically engineered T cells become a more significant focus of clinical investigation, identification of patient-specific neoantigen-reactive T cell receptors (TCRs) in a practical and efficient manner is a top priority. Using high-dimensional single-cell analysis, we recently identified a common gene expression signature in anti-tumor, neoantigen-reactive tumor-infiltrating lymphocytes (TIL) from patients with metastatic cancer, which included high gene expression of cell-surface markers of exhaustion. 1 Furthermore, analyses of intra-tumoral T cell populations have shown that neoantigen-specific TIL are enriched in subsets defined by cell-surface markers of exhaustion, likely secondary to oligoclonal expansion that occurs upon tumor antigen recognition in vivo. 2–5 In this study we describe an efficient method to prospectively capture and reconstruct neoantigen-reactive T cell receptors from tumor digest based on co-expression of CD39, PD-1, and TIGIT. Methods We evaluated the ability of PD-1+, CD39+, and TIGIT+ TIL (TIL-TP) to enrich for neoantigen-reactivity by sorting, sequencing, and reconstructing high-frequency TCR alpha/beta pairs from tumor digest in 5 patients with metastatic epithelial cancers. We then tested the ability of PD-1/CD39/TIGIT co-expressing TILs to sustain reactivity to patient-specific tumor neoantigens following in vitro expansion under similar conditions to our current clinical trial protocols (figure 1). Results We prospectively reconstructed TIL-TP TCRs to identify additional novel TCRs, with 35% of prospectively screened TCRs being neoantigen- or tumor-reactive. Including both previously known and newly predicted TCRs, TIL-TP demonstrated enrichment for neoantigen-reactivity in 4 of 5 patients, with a median of at least 26.8% of sequenced TIL-TP cells being neoantigen-reactive (range: 11.9 – 88.4%). TIL-TP TCR isolation demonstrated a high degree of correlation with single-cell transcriptomic approaches to identification of neoantigen-reactive TCRs, though TIL-TP TCRs represent a more cost-effective and widely available approach compared to those utilizing more advanced technologies. However, despite their substantial enrichment for neoantigen-reactive TCR clonotypes, the majority of TIL-TP populations failed to demonstrate neoantigen-reactivity following in vitro expansion and exhibited loss of neoantigen-reactive clones as well as functional impairment. Abstract 167 Figure 1 Conclusions TIL-TP serve as a highly efficient and reliable source of tumor-reactive TCRs. While direct utilization of these TIL-TP as a source for cellular therapy presents significant challenges, sorting for TIL-TP offers a streamlined approach using readily available and affordable technology to identify neoantigen-reactive TCRs that may be used to design TCR engineered cellular immunotherapies. References Lowery, FJ, et al. Single cell mapping of tumor infiltrating lymphocytes enables neoantigen-reactive T cell identification in metastatic human cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10–15 and May 17–21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 127. Duhen, T. et al. Co-expression of CD39 and CD103 identifies tumor-reactive CD8 T cells in human solid tumors. Nat Commun 2018; 9 :2724, doi:10.1038/s41467-018-05072-0. Gros A, et al. PD-1 identifies the patient-specific CD8(+) tumor-reactive repertoire infiltrating human tumors. J Clin Invest 2014; 124 :2246–2259, doi:10.1172/JCI73639. Pasetto, A. et al. Tumor- and Neoantigen-Reactive T-cell Receptors Can Be Identified Based on Their Frequency in Fresh Tumor. Cancer Immunol Res 2016; 4 :734–743, doi:10.1158/2326-6066.CIR-16-0001. Yossef, R. et al. Enhanced detection of neoantigen-reactive T cells targeting unique and shared oncogenes for personalized cancer immunotherapy. JCI Insight 2018; 3 : doi:10.1172/jci.insight.122467.
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2021
    detail.hit.zdb_id: 2719863-7
    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...