scholarly journals Targeted T-cell Therapy in Stage IV Breast Cancer: A Phase I Clinical Trial

2015 ◽  
Vol 21 (10) ◽  
pp. 2305-2314 ◽  
Author(s):  
Lawrence G. Lum ◽  
Archana Thakur ◽  
Zaid Al-Kadhimi ◽  
Gerald A. Colvin ◽  
Francis J. Cummings ◽  
...  
2016 ◽  
Vol 39 (3) ◽  
pp. 140-148 ◽  
Author(s):  
Hyeon-Seok Eom ◽  
Beom K. Choi ◽  
Youngjoo Lee ◽  
Hyewon Lee ◽  
Tak Yun ◽  
...  

PLoS ONE ◽  
2014 ◽  
Vol 9 (1) ◽  
pp. e83786 ◽  
Author(s):  
Takeshi Ishikawa ◽  
Satoshi Kokura ◽  
Tatsuji Enoki ◽  
Naoyuki Sakamoto ◽  
Tetsuya Okayama ◽  
...  

2018 ◽  
Vol 36 (15_suppl) ◽  
pp. 3019-3019 ◽  
Author(s):  
Stacey L. Doran ◽  
Sanja Stevanovic ◽  
Sabina Adhikary ◽  
Jared J. Gartner ◽  
Li Jia ◽  
...  

2014 ◽  
Vol 32 (15_suppl) ◽  
pp. 615-615
Author(s):  
Mary L. Disis ◽  
Andrew L Coveler ◽  
Doreen Higgins ◽  
Leonard A D'Amico ◽  
Chihiro Morishima ◽  
...  

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1797-1797 ◽  
Author(s):  
Jae H. Park ◽  
Isabelle Rivière ◽  
Xiuyan Wang ◽  
Jolanta Stefanski ◽  
Qing He ◽  
...  

Abstract Abstract 1797 Patient T cells may be genetically modified to express chimeric antigen receptors (CARs) targeted to antigens expressed on tumor cells. We have previously reported initial results from a phase I clinical trial treating patients with chemotherapy refractory chronic lymphocytic leukemia (CLL) with autologous T cells modified to express the 19–28z CAR targeted to the CD19 antigen expressed on most B cell malignancies (Brentjens RJ, Rivière I et al., Blood, 2011;118(18):4817-28). In the previous reported cohorts of 8 patients, CAR-modified T cells were infused in the setting of rapidly progressive and chemotherapy refractory disease. Although prior conditioning therapy with cyclophosphamide enhanced in vivo persistence of the modified T cells, all patients had cyclophosphamide-resistant disease and none experienced objective remissions or significant hematologic recovery. We hypothesized that suboptimal clinical response observed in the study was because of a large tumor burden at the time of T cell infusion and refractoriness to conditioning therapy. On the basis of these findings, we have modified the protocol to allow prior cytoreductive therapy and conditioning with chemotherapeutic agents based on predicted chemosensitivity. Since these protocol modifications, two patients have been treated. Both had relapsed disease with unfavorable disease phenotype following previous treatments with various chemotherapy and biologic regimens. Of the two patients treated to date, one achieved partial remission (PR) and the other attained minimal residual disease (MRD)-negative complete remission (CR) according to standard international criteria. The first patient experienced reduction in peripheral lymphocytosis and obtained stable disease with persistent anemia and thrombocytopenia after two cycles of bendamustine and rituximab (BR). Following the bendamustine conditioning and modified T cell infusion, PR was achieved with complete hematologic recovery, lasting for more than 8 months at the time of this report. The second patient achieved PR following two cycles of BR and subsequently attained MRD-negative CR with concomitant development of B cell aplasia after receiving the bendamustine conditioning and CAR-modified T cells. At the time of this report, the response has been sustained for more than 5 months. Notably, this patient has long-term persistence of the CAR-modified T cells, detected at 12 weeks following the T cell infusion. No significant toxicities were observed in the two patients, except for fevers lasting 3–4 days and transient grade 2 hypoxia. While the number of treated patients on the revised protocol is too small to draw a definitive conclusion, our findings of a significant improvement in the degree and depth of response with the bendamustine conditioning compared to our previous cohorts of cyclophosphamide-refractory CLL who received cyclophosphamide as their conditioning suggest a potentially greater effect of conditioning regimens through tumor burden reduction than the induction of a supportive cytokine response or lymphocyte depletion. In light of these initial observations, the role of the conditioning chemotherapy regimen given prior to adoptive T cell therapy needs to be carefully evaluated since not all regimens may ultimately be equally effective with respect to clinical outcomes. Disclosures: Lamanna: Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A482-A482
Author(s):  
Bryan Barnes ◽  
Ming Shan ◽  
Kristin Blouch ◽  
Mehmet Altan ◽  
Jaegil Kim ◽  
...  

BackgroundThis analysis evaluates an NY-ESO-1 immunohistochemistry (IHC) clinical trial assay in multiple tumor types for the identification of patients who may be eligible for NY-ESO-1 TCR T-cell targeted therapy. We provide an analysis of NY-ESO-1 expression and prevalence in non-small cell lung carcinoma (NSCLC) tumor samples from a patient cohort of an early Phase I/II clinical trial assessing NY-ESO-1 TCR T-cell therapy. Furthermore, we describe exploratory analyses of NY-ESO-1 prevalence and expression in a preliminary set of multiple tumor types to identify new indications for NY-ESO-1 TCR T-cell therapy.MethodsAn IHC assay was developed to detect NY-ESO-1 expression in formalin-fixed paraffin-embedded (FFPE) specimens utilizing an anti-NY-ESO-1 monoclonal antibody, clone E978. NY-ESO-1 protein expression levels and diagnostic status were determined by pathological evaluation under light microscopy to capture the percentage of tumor cell staining across all tumor cells in specimens at staining intensities 0, 1+, 2+ and 3+. NY-ESO-1 expression data were assessed for: prevalence using a ≥10% cutoff at ≥ 1+ intensity to assign positivity, and prevalence across classification (primary and metastatic) and subtype (adenocarcinoma and squamous cell carcinoma) for the NSCLC specimens.ResultsThe overall prevalence for NSCLC specimens from the Phase I/II trial was 15% (49/325) for NY-ESO-1. A prevalence of 15% (29/191) for primary and 14% (19/132) for metastatic samples, 13% (20/159) for adenocarcinoma, and 14% (5/35) for squamous cell carcinoma was observed. No significant difference was observed between subtype or%Tumor at each intensity. The preliminary set of indications used in exploratory studies had an observed prevalence as follows: gastric adenocarcinoma, 14 (4/28)%; esophageal adenocarcinoma & gastric esophageal junction, 9% (3/35); urothelial, 19% (6/31); head and neck squamous cell carcinoma, 10% (3/30); triple negative breast, 10% (3/30); hepatocellular carcinoma, 3%(1/30); and melanoma, 11% (3/27). NY-ESO-1 protein expression was localized in the cells’ nuclei and surrounding cytoplasm.ConclusionsMultiple indications assessed by the IHC clinical trial assay demonstrated similar NY-ESO-1 expression across the range of staining intensities and percentage of positive tumor observed as that in NSCLC, therefore warranting further development and validation of an IHC assay for NY-ESO-1 detection in these additional tumor types for use in clinical trials. These data support the use of IHC as a tool for the identification of patients whose tumors upregulate NY-ESO-1 in NSCLC and further encourage the investigation of multiple tumor types that may upregulate NY-ESO-1 as potential targets for NY-ESO-1 TCR T-cell therapies.AcknowledgementsThis study (NCT03709706) was funded by GlaxoSmithKline.Trial RegistrationNCT03709706ReferencesThomas R, et al. Front Immunol 2018;9:947Ethics ApprovalThis study was approved by the appropriate institutional review boards and independent ethics committees.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. TPS2619-TPS2619
Author(s):  
Marco L. Davila ◽  
Isabelle Riviere ◽  
Xiuyan Wang ◽  
Shirley Bartido ◽  
Jolanta Stefanski ◽  
...  

TPS2619 Background: Complete remission (CR) rates for B-ALL in adults is high (>80%), but overall survival remains low (approximately 30%). Patients with relapsed disease have a very poor prognosis with a median survival measured in months. Therefore, there is a dire need for novel therapies for adults with relapsed or minimal residual B-ALL. To this end we have developed a novel T cell therapy for B cell malignancies using patient derived T cells genetically modified to express the 19-28z chimeric antigen receptor (CAR), an artificial T cell receptor specific to the CD19 antigen expressed on most B cell tumors. Human T cells retrovirally modified to express the 19-28z CAR successfully targets and eradicates B cell tumors in vitro and systemic human B cell cancers in mice. We have recently published the results of our initial Phase I clinical trial experience (Brentjens, Rivière et al., Blood 2011) utilizing this technology in adults with chronic lymphocytic leukemia. These results suggest clinical activity and demonstrate a persistence of 19-28z+ T cells that is inversely associated with tumor burden. These studies suggest that 19-28z+ T cells may be particularly effective in relapsed B-ALL because these tumors retain a degree of chemotherapy sensitivity, which will allow the infusion of T cells after salvage chemotherapy when patients have low tumor burdens. To our knowledge, this is the first clinical trial using autologous CD19 targeted T cells in adults with B-ALL. Methods: We are enrolling patients to a Phase I T cell dose escalation trial (NCT01044069). Adults with CD19+ B-ALL classified as a CR, relapsed, or refractory are eligible for enrollment. After patients are enrolled they are leukapheresed to obtain peripheral blood T cells, which are then retrovirally transduced to express the 19-28z CAR and expanded in our GMP gene transfer facility. Enrolled patients with relapsed or refractory B-ALL undergo a cycle of re-induction chemotherapy and conditioning cyclophosphamide followed by infusion of autologous 19-28z+ T cells. To date, 11 patients have been enrolled and 2 patients have been infused with 19-28z+ T cells.


Sign in / Sign up

Export Citation Format

Share Document