scholarly journals Treatment of a cancer patient by an adoptive cell therapy protocol combining DC vaccination with cbl-b ex vivo silencing

2015 ◽  
Vol 3 (S2) ◽  
Author(s):  
Monika Sachet ◽  
Guenther Lametschwandtner ◽  
Hubert Hayden ◽  
Michaela Hassler ◽  
Hans Loibner ◽  
...  
2020 ◽  
Vol 04 (04) ◽  
pp. 345-350
Author(s):  
Ryan J. Slovak ◽  
Hyun S. Kim

AbstractThe reinfusion of autologous or allogeneic immune cells that have been educated and/or engineered ex vivo to respond to tumor-specific antigens is termed “adoptive cell therapy.” While adoptive cell therapy has made tremendous strides in the treatment of hematologic malignancies, its utilization for solid tumors has lagged somewhat behind. The purpose of this article is to concisely review the clinical research that has been done to investigate adoptive cell therapy as a treatment for gastrointestinal malignancies.


2021 ◽  
Vol 12 ◽  
Author(s):  
Ratchapong Netsrithong ◽  
Methichit Wattanapanitch

Adoptive cell therapy (ACT) using chimeric antigen receptor (CAR) T cells holds impressive clinical outcomes especially in patients who are refractory to other kinds of therapy. However, many challenges hinder its clinical applications. For example, patients who undergo chemotherapy usually have an insufficient number of autologous T cells due to lymphopenia. Long-term ex vivo expansion can result in T cell exhaustion, which reduces the effector function. There is also a batch-to-batch variation during the manufacturing process, making it difficult to standardize and validate the cell products. In addition, the process is labor-intensive and costly. Generation of universal off-the-shelf CAR T cells, which can be broadly given to any patient, prepared in advance and ready to use, would be ideal and more cost-effective. Human induced pluripotent stem cells (iPSCs) provide a renewable source of cells that can be genetically engineered and differentiated into immune cells with enhanced anti-tumor cytotoxicity. This review describes basic knowledge of T cell biology, applications in ACT, the use of iPSCs as a new source of T cells and current differentiation strategies used to generate T cells as well as recent advances in genome engineering to produce next-generation off-the-shelf T cells with improved effector functions. We also discuss challenges in the field and future perspectives toward the final universal off-the-shelf immunotherapeutic products.


2021 ◽  
Author(s):  
Sanghyun Kim ◽  
Nolan Vale ◽  
Nikolaos Zacharakis ◽  
Sri Krishna ◽  
Zhiya Yu ◽  
...  

Abstract Adoptive cell therapy (ACT) targeting neoantigens can achieve durable clinical responses in patients with cancer. Most neoantigens arise from rare mutations, requiring highly individualized treatments. To broaden the applicability of ACT targeting neoantigens, we focused on TP53 mutations commonly shared across different cancer types. Here, we describe a library of T cell receptors (TCRs) that can target TP53 mutations shared among 7.3% of patients with solid cancers. These TCRs recognized tumor cells in a TP53 mutation- and human leucocyte antigen (HLA)-specific manner both in vitro and in vivo. Patients with chemorefractory epithelial cancers treated with ex vivo-expanded autologous tumor infiltrating lymphocytes (TILs) naturally reactive with mutant p53 experienced limited clinical responses (2 PRs/12 patients), and we detected low frequencies, exhausted phenotypes, and poor persistence of the infused mutant p53-reactive TILs. Alternatively, we treated one patient with a chemorefractory breast cancer with ACT by transducing autologous peripheral blood lymphocytes with an HLA-A*02-restricted anti-p53R175H TCR. The infused cells exhibited an improved immunophenotype and prolonged persistence compared to the TIL ACT and the patient experienced an objective tumor regression (-55%) that lasted 6 months. Collectively, these data demonstrate the feasibility of off-the-shelf TCR-engineered cell therapies targeting shared p53 neoantigens to treat human cancers.


2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 12516-12516
Author(s):  
P. Pedrazzoli ◽  
I. Tunin ◽  
C. Tullio ◽  
E. Montini ◽  
R. Schiavo ◽  
...  

12516 Background: Ex-vivo generation of cytotoxic T cell (CTL) lines able to selectively kill autologous tumor cells (TC) is crucial for adoptive cell therapy of cancer. We have described the ex vivo generation of anti-tumor HLA-restricted CTL using CD8-enriched blood mononuclear cells (PBMC) and dendritic cells, pulsed with apoptotic TC as source of tumor antigens (Montagna, Int J Cancer 2004). These cells possess a marked cytotoxicity against TC but very low levels of cytotoxicity against autologous non-malignant controls, suggesting their safe in vivo use. Aim of this study was to evaluate the possibility of generating a large number of anti-tumor CTL lines, in compliance with good manufacture procedure (GMP), to be utilized for adoptive cell therapy. Methods: Four patients with advanced, pretreated solid tumors have been evaluated: 2 sarcoma, 1 renal carcinoma and 1 ovarian carcinoma. Tumor sample was obtained during surgical procedures planned for therapeutic purposes, while PBMC were obtained by a single apheresis. All reagents employed for cultures were produced under GMP. Based on our reported method, the current protocol included two or three rounds of tumor-specific stimulation, followed by two rounds of antigen-independent expansion. Results: We have obtained, after the first round of expansion and in total, a range of 0.5–2×109 and 5–20×109 CTL, respectively. Such large amounts of anti-tumor CTL were generated even if a low number of viable TC was available. In 3 out of 4 anti-tumor CTL lines, >85% of effector cells were CD3+/CD8+. Conclusions: We demonstrated the feasibility of obtaining, in GMP conditions, large quantities of anti-tumor specific CTL suitable for in vivo use. A phase I protocol of lymphoablative therapy followed by CTL transfer in vivo has been designed and submitted for approval to the competent regulatory agency. No significant financial relationships to disclose.


2013 ◽  
Vol 19 (8) ◽  
pp. 2132-2143 ◽  
Author(s):  
Yin Liu ◽  
Hong-Wei Wu ◽  
Michael A. Sheard ◽  
Richard Sposto ◽  
Srinivas S. Somanchi ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 929-929
Author(s):  
Matthew J Goldstein ◽  
Bindu Varghese ◽  
Ranjani Rajapaksa ◽  
Joshua Brody ◽  
Shoshana Levy ◽  
...  

Abstract Abstract 929 Background: Recently, we have investigated adoptive cell therapy for treating lymphoma. The efficacy of this maneuver has been demonstrated by curing large established tumors. Specifically, we use active immunization to generate anti-tumor T cells in vivo and transfer these T cells into lymphodepleted recipient mice (Brody J, Goldstein MJ, Czerwinski DK, and Levy R; Blood, 2009). A major challenge in adoptive therapy is the method of generating anti-tumor T cells. Traditionally, tumor-specific T cells are expanded to large numbers ex vivo. Herein, we describe a new, whole-cell vaccine that is effective in inducing anti-tumor T cells in vivo. This vaccine combines tumor antigens with an immune stimulant: irradiated-tumor cells (a source of tumor antigens) are loaded with the TLR agonist CpG (an immune stimulant). Our vaccine approach has several potential advantages: (1) anti-tumor immunity generated by our CpG-loaded, whole-cell vaccine is poly-antigenic and thus, not limited by the expression of a single antigen target on tumor cells; (2) ex vivo expansion may generate large numbers of effector T cells that can induce tumor regression in the short-term, but have a limited ability to maintain a persistent anti-tumor response. Our model avoids ex vivo manipulation of anti-tumor T cells and thus, may preserve and enhance a memory T cell population that sustains the anti-tumor response. Methods: We derived a new pre-B cell lymphoma cell line in the C57BL/6 background. Primary bone marrow cells were isolated from C57BL/6 donor mice and transfected with a recombinant retrovirus containing the Bcr-Abl oncogene. The emerging transformed cell line was designated H11. This cell line expressed the B lineage marker CD19 but was negative for MHC II and surface Ig. Irradiated H11 tumor cells were pre-loaded with CpG for 24 hours and administered to donor mice by daily, sub-cutaneous injections for five days. Donor splenocytes were harvested seven days following vaccination and adoptively transferred into lethally irradiated recipient mice that were subsequently challenged with a lethal dose of H11 tumor cells. Results: Vaccination with CpG-loaded H11 tumor cells (CpG-H11) generated anti-tumor T cells that are effective in adoptive cell therapy. 100% of mice receiving adoptive therapy with vaccine-induced T cells were protected from tumor challenge. In contrast, vaccination of donor mice with untreated H11 tumor was insufficient for generating anti-tumor T cells. Only 20% of mice treated with T cells from these donors were protected from tumor challenge. In spite of the H11 tumor being MHC Class II−, we observed that anti-tumor immunity generated by the CpG-H11 vaccine was CD4 T cell mediated. CD4 T cells were isolated from CpG-H11 vaccinated donors by flow cytometry. Fewer than 1.8×106 CD4 T cells were sufficient to protect 80% of recipient mice from tumor challenge. In contrast, equivalent numbers of donor CD8 T cells provided no benefit. These results strongly suggest that the CpG-H11 vaccine induced cross-presentation of tumor antigens by antigen-presenting cells (APCs). We have demonstrated that CpG-loaded H11 tumor cells can leak CpG into the immediate environment activating nearby APCs. These APCs have greater phagocytic potential and express higher levels of co-stimulatory molecules such as CD40. Ongoing studies will determine whether APCs which encounter the CpG-H11 vaccine but not untreated H11 tumor cells, can stimulate proliferation of anti-tumor T cells. Conclusions: Here we describe a novel, whole-cell vaccine approach that induces anti-tumor T cells for adoptive therapy to treat lymphoma. This vaccine is superior to vaccination with tumor cells alone. We are currently developing this therapy for evaluation in a clinical trial to treat mantle cell lymphoma. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 13 ◽  
pp. 175883592110083
Author(s):  
Apostolos Sarivalasis ◽  
Matteo Morotti ◽  
Arthur Mulvey ◽  
Martina Imbimbo ◽  
George Coukos

Epithelial ovarian cancer (EOC) is the most important cause of gynecological cancer-related mortality. Despite improvements in medical therapies, particularly with the incorporation of drugs targeting homologous recombination deficiency, EOC survival rates remain low. Adoptive cell therapy (ACT) is a personalized form of immunotherapy in which autologous lymphocytes are expanded, manipulated ex vivo, and re-infused into patients to mediate cancer rejection. This highly promising novel approach with curative potential encompasses multiple strategies, including the adoptive transfer of tumor-infiltrating lymphocytes, natural killer cells, or engineered immune components such as chimeric antigen receptor (CAR) constructs and engineered T-cell receptors. Technical advances in genomics and immuno-engineering have made possible neoantigen-based ACT strategies, as well as CAR-T cells with increased cell persistence and intratumoral trafficking, which have the potential to broaden the opportunity for patients with EOC. Furthermore, dendritic cell-based immunotherapies have been tested in patients with EOC with modest but encouraging results, while the combination of DC-based vaccination as a priming modality for other cancer therapies has shown encouraging results. In this manuscript, we provide a clinically oriented historical overview of various forms of cell therapies for the treatment of EOC, with an emphasis on T-cell therapy.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. 3045-3045 ◽  
Author(s):  
Amod Sarnaik ◽  
Harriet M. Kluger ◽  
Jason Alan Chesney ◽  
Jyothi Sethuraman ◽  
Anandharamen Veerapathran ◽  
...  

3045 Background: Adoptive cell therapy with TIL involves collection of autologous lymphocytes from the tumor via surgical resection, ex vivo expansion of TIL, lymphodepletion of the patient prior to infusion of TIL using Fludarabine and Cyclophosphamide, followed by infusion of TIL. Up to 6 doses of IL-2 (600,000 IU/kg) is administered to support multiplication of TIL and engraftment. Here, we present the preliminary results from an ongoing, multi-site Phase 2 study of TIL for advanced metastatic melanoma. Methods: Patients with advanced metastatic melanoma who have failed at least one prior systemic therapy were enrolled. Primary objective of the study was to characterize safety profile of LN-144. At baseline, patients had a median age of 56 (41-72) years; 44% were ≥ 60 years old. Median sum of tumor diameters for the target lesions was 10.4 cm, and median of 3 prior therapies. All enlisted patients had prior anti-PD1 as well as anti-CTLA4 and 67% had received ≥ 3 prior therapies. Responses were assessed by RECIST 1.1. TIL products were centrally manufactured. No complications arose from shipment of tumors or TIL. Results: Results are presented through 31 Jan 2017 for the first 9 infused patients evaluable by two assessments. Eight of 9 patients received all 6 doses of IL-2 per protocol. The most common (≥3 patients) non-hematologic grade 3-4 TEAE was hypophosphatemia. No neurotoxicity of grade ≥ 3 was reported. There were no deaths or discontinuations due to SAEs related to study treatment. ORR was 33% (CR = 11%, PR = 22%, SD = 22%, PD = 33%, NE = 11%). Mean time to best response was 3.0 months and median duration of follow up was 3.6 months (1.1+, 12.1). Responses were observed in patients with tumors carrying wild type or BRAF mutations. All patients demonstrated persistence of TIL on day 14 post-infusion. Conclusions: Cell therapy with TIL is an effective treatment with acceptable safety profile for advanced metastatic melanoma patients who are refractory to anti-PD1. TIL products can be centrally manufactured for broad clinical application. This study will be expanded to enroll patients with a shorter manufacturing process as well as offering retreatment for study patients. Clinical trial information: NCT02360579.


2014 ◽  
Vol 21 (3) ◽  
pp. 611-621 ◽  
Author(s):  
Jessica Ann Chacon ◽  
Amod A. Sarnaik ◽  
Jie Qing Chen ◽  
Caitlin Creasy ◽  
Charuta Kale ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document