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
Filter
  • American Association for Cancer Research (AACR)  (9)
  • 1
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 11, No. 12_Supplement ( 2023-12-01), p. A042-A042
    Abstract: The Pediatric Leukemia Adoptive Therapy (PLAT) trials at Seattle Children’s Hospital have pioneered the use of chimeric antigen receptor (CAR) T cell (CAR-T) therapies in pediatric patients with B cell Acute Lymphoblastic Leukemia. The landmark PLAT-02 trial (n=45), utilizing a second-generation CD19-directed CAR, observed a 93% remission rate following CAR-T infusion. However, approximately half of these subjects relapsed within one year and nearly a third of those tumors no longer expressed the CD19 target. The PLAT-04 trial (n=4) was subsequently conceived with a second-generation CAR (SCRI-C22v1) engineered to target CD22, an alternative antigen expressed on the B cell surface. Despite promising preclinical data, SCRI-C22v1 failed to generate anti-leukemic activity in all four subjects and the trial was halted. In response, a different second-generation CD22-directed CAR (SCRI-C22v2) was engineered, replacing the CD28 transmembrane domain and IgG4 hinge domain in SCRI-C22v1 with a continuous CD8 hinge and transmembrane domain while retaining identical 4-1BB and CD3ζ intracellular signaling domains. The modified SCRI-C22v2 displayed improved anti-tumor activity in preclinical models and was ultimately tested in patients in the PLAT-07 trial (n=4), where it exhibited strong in vivo expansion and persistence while inducing complete remission after 1 month in all four subjects. To investigate the molecular signaling responsible for clinical differences observed between SCRI-C22v1 and SCRI-C22v2, we applied an emerging proteomic technology, Quantitative Multiplex Immunoprecipitation (QMI), that captures medium-throughput quantitative data about fold changes in protein interaction networks downstream of the CAR. CAR-T products from healthy donors expressing either SCRI-C22v1 or SCRI-C22v2 were stimulated with fixed K562 cells expressing CD22 and protein interaction networks were profiled with QMI, revealing distinct differences in signal transduction downstream of the CAR. Contrary to expectation, it was the ineffective SCRI-C22v1 that exhibited significantly enhanced signalosome formation, engaging more strongly than SCRI-C22v2 with both the classical CD3ζ-ZAP70-LAT-SLP76 pathway as well as the non-classical TRAF pathway. Interestingly, the clinically successful SCRI-C22v2 demonstrated stronger baseline engagement with inhibitory molecules that attenuate canonical TCR signaling such as SHP2 and UBASH3A, suggesting regulatory control of CAR signaling is crucial for optimal clinical responses. Since the CAR is a synthetic receptor, it should be possible to bioengineer optimized signal transduction to some extent, and our group continues to work towards identifying an optimized QMI signature that can be used as a CAR-tune network for the rational design of more effective CAR-T therapies. Citation Format: Eric Bueter, Corinne Summers, Isabella Draper, Joshua Gustafson, Stephen EP Smith, Kamila Gwiazda, Julie Park, Colleen Annesley, Rebecca Gardner, Michael Jensen. Quantitative multiplex immunoprecipitation reveals distinct protein interaction networks responsible for discrepant in vivo activity exhibited by two second generation CD22-targeted chimeric antigen receptors [abstract]. In: Proceedings of the AACR Special Conference in Cancer Research: Tumor Immunology and Immunotherapy; 2023 Oct 1-4; Toronto, Ontario, Canada. Philadelphia (PA): AACR; Cancer Immunol Res 2023;11(12 Suppl):Abstract nr A042.
    Type of Medium: Online Resource
    ISSN: 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 28, No. 18_Supplement ( 2022-09-15), p. IA020-IA020
    Abstract: Rhabdomyosarcoma, a pediatric malignancy with partial resemblance to undifferentiated skeletal muscle, is characterized by high expression of myogenic-lineage transcription factors such as MYOD1 and MYOG. Despite high expression of these transcription factors, which in normal muscle result in differentiation, RMS cells fail to differentiate, suggesting the presence of factors that inhibit their normal differentiation-promoting functions. In this talk, I will present data that the key muscle transcriptional regulator, SIX1, which in development activates the myogenic regulatory factors (MRFs) and promotes muscle differentiation, in fact inhibits differentiation in fusion-negative (FN) RMS. SIX1 holds FN-RMS cells in a progenitor-like state by altering the chromatin landscape and causing MYOD1, a key MRF, to preferentially bind to regulatory regions of genes permissive to growth rather than differentiation. Loss of SIX1 results in re-localization of MYOD1 to promoters/enhancers of genes associated with differentiation, and further results in increased binding of MYOG at such loci. Altered binding of MYOD1 and MYOG in response to SIX1 loss results in marked inhibition of RMS growth in vivo, via induction of differentiation. These data suggest that SIX1 acts as a master regulatory factor controlling the fate of RMS cells. Data will be presented that suggest dynamic actions of SIX1 and its co-factors throughout normal muscle differentiation, whereby high levels of SIX1 expressed in early muscle differentiation may mimic a transcriptional state seen in RMS. We hypothesize that the specific levels of SIX1, combined with a unique combination of transcriptional co-factors, reprogram genome-wide binding of MRFs to different promoter/enhancer sites at specific developmental time points, and that RMS is trapped in an early developmental state where SIX1 represses differentiation via genome-wide alterations in MRF binding that favor growth. Understanding the co-factors that work with SIX1 to alter chromatin state and MRF binding may enable the discovery of novel targets whose inhibition could serve as a relatively non-toxic treatment to restore normal developmental processes and inhibit RMS progression. Citation Format: Heide L. Ford, Jessica Y. Hsu, Etienne P. Danis, Stephanie Nance, Jenean H. O’Brien, Annika L. Gustafson, Veronica M. Wessells, Andrew E. Goodspeed, Jared C. Talbot, Sharon L. Amacher, Paul Jedlicka, Joshua C. Black, James C. Costello, Adam D. Durbin, Kristin B. Artinger. Reprogramming of myogenic transcription factors in rhabdomyosarcoma [abstract]. In: Proceedings of the AACR Special Conference: Sarcomas; 2022 May 9-12; Montreal, QC, Canada. Philadelphia (PA): AACR; Clin Cancer Res 2022;28(18_Suppl):Abstract nr IA020.
    Type of Medium: Online Resource
    ISSN: 1557-3265
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 21 ( 2019-11-01), p. 5652-5667
    Abstract: MYCN is a major driver for the childhood cancer, neuroblastoma, however, there are no inhibitors of this target. Enhanced MYCN protein stability is a key component of MYCN oncogenesis and is maintained by multiple feedforward expression loops involving MYCN transactivation target genes. Here, we reveal the oncogenic role of a novel MYCN target and binding protein, proliferation-associated 2AG4 (PA2G4). Chromatin immunoprecipitation studies demonstrated that MYCN occupies the PA2G4 gene promoter, stimulating transcription. Direct binding of PA2G4 to MYCN protein blocked proteolysis of MYCN and enhanced colony formation in a MYCN-dependent manner. Using molecular modeling, surface plasmon resonance, and mutagenesis studies, we mapped the MYCN–PA2G4 interaction site to a 14 amino acid MYCN sequence and a surface crevice of PA2G4. Competitive chemical inhibition of the MYCN–PA2G4 protein–protein interface had potent inhibitory effects on neuroblastoma tumorigenesis in vivo. Treated tumors showed reduced levels of both MYCN and PA2G4. Our findings demonstrate a critical role for PA2G4 as a cofactor in MYCN-driven neuroblastoma and highlight competitive inhibition of the PA2G4-MYCN protein binding as a novel therapeutic strategy in the disease. Significance: Competitive chemical inhibition of the PA2G4–MYCN protein interface provides a basis for drug design of small molecules targeting MYC and MYCN-binding partners in malignancies driven by MYC family oncoproteins.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    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 ...
  • 4
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 19, No. 8 ( 2020-08-01), p. 1598-1612
    Abstract: Since the discovery of CHD1L in 2008, it has emerged as an oncogene implicated in the pathology and poor prognosis of a variety of cancers, including gastrointestinal cancers. However, a mechanistic understanding of CHD1L as a driver of colorectal cancer has been limited. Until now, there have been no reported inhibitors of CHD1L, also limiting its development as a molecular target. We sought to characterize the clinicopathologic link between CHD1L and colorectal cancer, determine the mechanism(s) by which CHD1L drives malignant colorectal cancer, and discover the first inhibitors with potential for novel treatments for colorectal cancer. The clinicopathologic characteristics associated with CHD1L expression were evaluated using microarray data from 585 patients with colorectal cancer. Further analysis of microarray data indicated that CHD1L may function through the Wnt/TCF pathway. Thus, we conducted knockdown and overexpression studies with CHD1L to determine its role in Wnt/TCF-driven epithelial-to-mesenchymal transition (EMT). We performed high-throughput screening (HTS) to identify the first CHD1L inhibitors. The mechanism of action, antitumor efficacy, and drug-like properties of lead CHD1L inhibitors were determined using biochemical assays, cell models, tumor organoids, patient-derived tumor organoids, and in vivo pharmacokinetics and pharmacodynamics. Lead CHD1L inhibitors display potent in vitro antitumor activity by reversing TCF-driven EMT. The best lead CHD1L inhibitor possesses drug-like properties in pharmacokinetic/pharmacodynamic mouse models. This work validates CHD1L as a druggable target and establishes a novel therapeutic strategy for the treatment of colorectal cancer.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. LB-109-LB-109
    Abstract: Personalized immunotherapy has reached a pivotal milestone with the recent approval of CD19-directed chimeric antigen receptor (CAR) T-cell therapies for certain B-cell malignancies. Other than on-target off-tumor autoimmunity, the commonly seen toxicity is severe cytokine release syndrome (sCRS) which is somewhat managed with tocilizumab and corticosteroids. Although the landscape of CAR-T cell development includes & gt;200 clinical trials in various tumor types, success against solid tumors remains a significant challenge. Much of the unmet medical need has been attributed to limited “ideal” tumor-specific targets, tumor heterogeneity, CAR-T cell infiltration/survival, and the presence of an immunosuppressive tumor microenvironment. Due to frequent overexpression in many human cancers, the folate receptor (FR) has become an exploratory target for anti-FRα/β CAR-T cell therapies. We are developing a CAR-T adaptor molecule (CAM)-based therapy that uses the small-molecule bispecific ligand, EC17 (folate-FITC). In this setting, EC17 effectively “paints” FR+ tumors with the FITC antigen to attract anti-FITC CAR-T cells and induce an immune response. As previously confirmed in mice as well as in human clinical studies, EC17 penetrates solid tumors within minutes and is retained due to high affinity for the FR; whereas unbound EC17 rapidly clears from the blood and from receptor-negative tissues. When tested against human xenografts, EC17/CAR-T therapy showed consistent antitumor activity with low or no adverse reactions. However, sCRS was observed under defined experimental conditions. Further testing revealed that sCRS of grade ≥3 (out of a 0-5 scale) can be mitigated or even prevented using intermittent “on-and-off” dosing and/or dose titration of the EC17 CAM. But, under extreme conditions where dose cessation failed, we discovered that intravenous sodium fluorescein (NaFl) could be used as a fast-acting rescue agent to temporarily displace CAR-T cells from their targets and reverse the sCRS symptoms. For translation into first-in-human testing, we selected a high-affinity anti-FITC CAR construct comprising (i) a humanized FITC scFv (E2; 0.75 nM Kd), (ii) an IgG4 hinge-CH2-CH3 spacer fused to a CD28-transmemeber domain, (iii) a second-generation 4-1BB:zeta-endodomain, and (iv) a cell-surface human EGFRt tag. Co-culture experiments were performed using this construct to establish EC17-dependent dose response and confirm target specificity as well as FR expression threshold using a panel of FR-positive cancer cell lines. Clinically relevant EC17 dosing regimens were evaluated using tumor-free and tumor-bearing mice to study CAR-T cell proliferation, cytokine production and the onset/mitigation of sCRS. We confirmed that EC17 administration in the presence of FR+ tumors was the key to drive in vivo CAR-T cell proliferation towards a more persistent phenotype. In addition, higher levels of CAR-T cells were detected in metastatic tumors versus adjacent normal tissues. Finally, EC17/CAR-T cell therapy demonstrated remarkable efficacy against some of the more aggressive and chemo-resistant FR+ tumors of various histology. In summary, our CAM-driven CAR-T cell approach provides antitumor activity with multiple safety control mechanisms. Citation Format: Yingjuan June Lu, Haiyan Chu, Leroy W. Wheeler, Mellissa Nelson, Elaine M. Westrick, Marilynn R. Vetzel, Patrick J. Klein, Adam J. Johnson, Jason K. Yokoyama, Joshua A. Gustafson, Michael C. Jensen, Yong-Gu Lee, Philip S. Low, Christopher P. Leamon. Adaptor controlled CAR-T cell immunotherapy for treatment of folate receptor-alpha/beta positive solid and liquid tumors [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr LB-109.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Research Vol. 76, No. 14_Supplement ( 2016-07-15), p. 1522-1522
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 1522-1522
    Abstract: The increasing availability of genetic data has led to a demand for robust methods to extract information and an increased need for complex computational analysis. The DREAM project consisted of 44 teams assessing various computational methods to predict drug sensitivity in 53 breast cancer cell lines. (Nat Biotechnol 32:1202). Results from this and other studies suggest that gene expression data from microarrays are one of the more valuable and robust data sets for predicting drug sensitivity. Whole exome sequencing is a valuable tool for predicting sensitivity to targeted agents, however the mutational spectrum is not as predictive for cytotoxic agents (Cancer Res 73:4372). Thus, a systematic analysis of the predictive ability of gene expression on cytotoxic chemotherapy drug sensitivity (IC50 values) using multiple cell line panels is an important component to understanding the versatility and applicability of gene expression for predicting cytotoxic drug IC50 values in tumor cells. Using GDSC and NCI60 cell panels, linear regression models were constructed to predict IC50 values from array data for 13 cytotoxic agents. To address the limited amount of data compared to a possible feature space & gt; 22,000 genes, the methods of Principle Least Squares Regression (PLSR) and Principle Component Regression (PCR) were employed. Models were generated using four different training and validating scenarios to evaluate performance. Mean Absolute Differences (MAD) achieved between expected and predicted IC50 values as low as 11% and correlations as large as 0.7 (P & lt;0.05) were achieved for some drugs. However, several obstacles have presented themselves. Mainly, among 35 cell lines shared by the GDSC and NCI60 direct correlation across common cell lines averaged only 0.4 highlighting the challenge to obtain, evaluate, and address consistent data suitable for robust models. Model Performance Metrics showing high, average, and low values across all thirteen drugs.ModelMeasurementPCR Low : Avg. : HighPLSR Low : Avg. : HighGDSC: TrainCorrelation0.16 : 0.36 : 0.70.12 : 0.36 : 0.66GDSC: ValidP-Value0a : 0.008 : 0.0090 : 0.02 : 0.16MAD% Range11 : 13 : 1611 : 13 : 16Classification Accuracy0.39 : 0.66 : 0.94NCI60: TrainCorrelation-0.11 : 0.36 : 0.67-0.21 : 0.4 : 0.79NCI60: ValidP-Value0.007 : 0.31 : 0.7550.0007 : 0.24 : 0.87MAD% Range11 : 17 : 2711 : 17 : 29Classification Accuracy0.33 : 0.62 : 0.75N/AGDSC: TrainCorrelation0.2 : 0.36 : 0.550.16 : 0.34 : 0.56NCI60: ValidP-Value0 : 0.03 : 0.130 : 0.06 : 0.23MAD% Range11 : 23 : 3711 : 23 : 39Classification Accuracy0.32 : 0.59 : 0.82N/ANCI60: TrainCorrelation0.023 : 0.19 : 0.48-0.03 : 0.18 : 0.43GDSC: ValidP-Value0 : 0.08 : 0.750 : 0.09 : 0.72MAD% Range12 : 19 : 3612 : 19 : 35Classification Accuracy0.37 : 0.53 : 0.69NAa Low p values of 0 correspond to p values & lt; 10−5 In summary, PCR and PLSR based gene expression models predict tumor cell sensitivity to some cytotoxic drugs with varying degrees of accuracy. Citation Format: Joshua Mannheimer, Jared S. Fowles, Katherine Shaumberg, Dawn L. Duval, Ashok Prasad, Daniel L. Gustafson. Predicting drug sensitivity based on gene array data for cytotoxic chemotherapeutic agents. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1522.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    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
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 26, No. 17 ( 2020-09-01), p. 4633-4642
    Abstract: The purpose of this study was to evaluate the rational combination of TORC1/2 inhibitor TAK-228 and Aurora A kinase inhibitor alisertib in preclinical models of triple-negative breast cancer (TNBC) and to conduct a phase I dose escalation trial in patients with advanced solid tumors. Experimental Design: TNBC cell lines and patient-derived xenograft (PDX) models were treated with alisertib, TAK-228, or the combination and evaluated for changes in proliferation, cell cycle, mTOR pathway modulation, and terminal cellular fate, including apoptosis and senescence. A phase I clinical trial was conducted in patients with advanced solid tumors treated with escalating doses of alisertib and TAK-228 using a 3+3 design to determine the maximum tolerated dose (MTD). Results: The combination of TAK-228 and alisertib resulted in decreased proliferation and cell-cycle arrest in TNBC cell lines. Treatment of TNBC PDX models resulted in significant tumor growth inhibition and increased apoptosis with the combination. In the phase I dose escalation study, 18 patients with refractory solid tumors were enrolled. The MTD was alisertib 30 mg b.i.d. days 1 to 7 of a 21-day cycle and TAK-228 2 mg daily, continuous dosing. The most common treatment-related adverse events were neutropenia, fatigue, nausea, rash, mucositis, and alopecia. Conclusions: The addition of TAK-228 to alisertib potentiates the antitumor activity of alisertib in vivo, resulting in increased cell death and apoptosis. The combination is tolerable in patients with advanced solid tumors and should be evaluated further in expansion cohorts with additional pharmacodynamic assessment.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 5722-5722
    Abstract: A key goal in immuno-oncology is the identification of tumor antigens recognized by CD8 T cells. Immune modulators such as PD-1 inhibitors indirectly promote T-cell attack against tumor antigens, and may be augmented by antigen-directed therapeutic immunization or adoptive cell therapy to increase clinical benefit. Such personalized therapeutics require accurate antigen identification from patient samples, which remains elusive today. Methods: We generated the largest reported dataset of human tumor transcriptomes and HLA class I peptidomes (N=111) from specimens of multiple tumor types. We used these data to train a deep learning model of HLA peptide presentation for antigen prediction. Our model architecture addressed two key challenges: (1) learning HLA-allele-specific models from tumor data where each sample expressed up to 6 unique HLA class I alleles and (2) incorporating information about all aspects of HLA presentation, including gene expression, proteasomal processing and stable binding of peptides to HLA. We evaluated the performance of the model on two independent test datasets. First, we tested the model on HLA presented peptides from five held-out tumor mass spectrometry samples. Then, to establish that accurate prediction of HLA presentation translates to prediction of antigens recognized by T cells in vivo, we compiled from three published studies a dataset of & gt;2,000 mutations in 16 patients, with 23 mutations (i.e. neoantigens) recognized by PD1+ PBMC or TIL CD8 T cells. Recognition of a peptide by TIL or activated peripheral T-cells implies not only tumor presentation of the peptide, but also its ability to prime T cell responses, and thus represents the most stringent test of tumor antigen prediction. Results: The model demonstrated a breakthrough in prediction accuracy. On the mass spectrometry test data, it achieved a & gt;10-fold improvement in positive predictive value (PPV) vs standard HLA binding affinity (~50% PPV at 40% recall for the MS-based model vs ~5% PPV for binding affinity prediction). On the T-cell test dataset, the model ranked T-cell recognized neoantigens on average & gt;4-fold higher than standard prediction (median rank ~7 for the MS-based model vs ~30 for binding affinity). When selecting candidate neoantigens for hypothetical 10-neoantigen personalized immunotherapy, the model prioritized at least 1 T-cell recognized neoantigen in the top 10 for 9/11 patients with neoantigen responses vs 3/11 for binding affinity. For a hypothetical 20-neoantigen immunotherapy, the model correctly selected the majority (16/23, 70%) of recognized neoantigens. Conclusion: We used the largest dataset of tumor transcriptomes and HLA peptidomes reported to-date to train a deep learning model of HLA epitope presentation. The new model significantly outperforms state of the art methods, and has sufficient predictive accuracy for in silico antigen selection for personalized cancer immunotherapy. Citation Format: Brendan Bulik-Sullivan, Jennifer Busby, Matthew Davis, Lauren Young, Tyler Murphy, Andrew Clark, Fujiko Duke, Michele Busby, Adnan Derti, Mojca Skoberne, Karin Jooss, Corinne E. Gustafson, Assunta De Rienzo, William G. Richards, Nhien T. Dao, Hyeong R. Kim, Jamie E. Anderson, Chang-Min Choi, Vincent De Montpreville, Se Jin Jang, Olaf Mercier, Raphael Bueno, Elie Fadel, Joshua Francis, Roman Yelensky. Antigen identification for cancer immunotherapy by deep learning on tumor HLA peptides [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5722.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    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 ...
  • 9
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 13, No. 1 ( 2023-01-09), p. 114-131
    Abstract: Diffuse intrinsic pontine glioma (DIPG) remains a fatal brainstem tumor demanding innovative therapies. As B7-H3 (CD276) is expressed on central nervous system (CNS) tumors, we designed B7-H3–specific chimeric antigen receptor (CAR) T cells, confirmed their preclinical efficacy, and opened BrainChild-03 (NCT04185038), a first-in-human phase I trial administering repeated locoregional B7-H3 CAR T cells to children with recurrent/refractory CNS tumors and DIPG. Here, we report the results of the first three evaluable patients with DIPG (including two who enrolled after progression), who received 40 infusions with no dose-limiting toxicities. One patient had sustained clinical and radiographic improvement through 12 months on study. Patients exhibited correlative evidence of local immune activation and persistent cerebrospinal fluid (CSF) B7-H3 CAR T cells. Targeted mass spectrometry of CSF biospecimens revealed modulation of B7-H3 and critical immune analytes (CD14, CD163, CSF-1, CXCL13, and VCAM-1). Our data suggest the feasibility of repeated intracranial B7-H3 CAR T-cell dosing and that intracranial delivery may induce local immune activation. Significance: This is the first report of repeatedly dosed intracranial B7-H3 CAR T cells for patients with DIPG and includes preliminary tolerability, the detection of CAR T cells in the CSF, CSF cytokine elevations supporting locoregional immune activation, and the feasibility of serial mass spectrometry from both serum and CSF. This article is highlighted in the In This Issue feature, p. 1
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2607892-2
    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...