scholarly journals IDO and galectin-3 hamper the ex vivo generation of clinical grade tumor-specific T cells for adoptive cell therapy in metastatic melanoma

2017 ◽  
Vol 66 (7) ◽  
pp. 913-926 ◽  
Author(s):  
Sara M. Melief ◽  
Marten Visser ◽  
Sjoerd H. van der Burg ◽  
Els M. E. Verdegaal
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.


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.


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.


2020 ◽  
Vol 8 (1) ◽  
pp. e000166 ◽  
Author(s):  
Els Verdegaal ◽  
Monique K van der Kooij ◽  
Marten Visser ◽  
Caroline van der Minne ◽  
Linda de Bruin ◽  
...  

BackgroundAdoptive cell therapy (ACT) with tumor-reactive T cells has shown consistent clinical efficacy. We evaluated the response to ACT in combination with interferon alpha (IFNa) preconditioning in patients with stage IV metastatic melanoma, most of which were progressive on cytotoxic T-lymphocyte-associated protein 4 and/or programmed cell death protein 1 checkpoint blockade therapy.MethodsThirty-four patients were treated with ex vivo expanded tumor reactive T cells, derived from mixed lymphocyte autologous tumor cultures, or with autologous tumor-infiltrating lymphocytes and evaluated for clinical response. Clinical and immunological parameters associated with response were also evaluated.ResultsBest overall response defined as clinical benefit, comprising either complete response, partial response or stable disease >6 months, was observed in 29% of the patients. Forty-three per cent of the 14 immunotherapy-naïve patients and 20% of the 20 patients progressive on prior immunotherapy benefited from ACT. The overall survival (OS) was 90% versus 28.6% at 1 year and 46.7% versus 0% at 3 years follow-up, of responder and non-responder patients, respectively. Median OS was 36 versus 7 months, respectively. IFNa pretreatment resulted in leukopenia, neutropenia and lymphopenia, which was sustained during the treatment in clinical responders and associated with response. Differences in antigen specificity, but not in phenotype, cytokine profile or CD8+ T cell number of the ACT products correlated with clinical response. Cross-reactivity of the ACT products to one or more allogeneic human leukocyte antigen-matched melanoma cell lines was associated with short OS after treatment while the ACT products of very long-term survivors showed no cross-reactivity but recognized patient-specific neoantigens.ConclusionThis study demonstrates that ACT in combination with a mild IFNa preconditioning regimen can induce clinical benefit even in immunotherapy pretreated patients, although with lower success than in immunotherapy-naïve patients. ACT products comprising neoantigen reactivity may be more effective.


2020 ◽  
Vol 47 (6) ◽  
pp. 464-471
Author(s):  
Lauren Suarez ◽  
Ruipeng Wang ◽  
Scott Carmer ◽  
Daniel Bednarik ◽  
Han Myint ◽  
...  

Over the last decade, tremendous progress has been made in the field of adoptive cell therapy. The two prevailing modalities include endogenous non-engineered approaches and genetically engineered T-cell approaches. Endogenous non-engineered approaches include dendritic cell-based systems and tumor-infiltrating lymphocytes (TIL) that are used to produce multi-antigen-specific T-cell products. Genetically engineered approaches, such as T-cell receptor engineered cells and chimeric antigen receptor T cells are used to produce single antigen-specific T-cell products. It is noted by the authors that there are alternative methods to sort for antigen-specific T cells such as peptide multimer sorting or cytokine secretion assay-based sorting, both of which are potentially challenging for broad development and commercialization. In this review, we are focusing on a novel nanoparticle technology that generates a non-engineered product from the endogenous T-cell repertoire. The most common approaches for ex vivo activation and expansion of endogenous, non-genetically engineered cell therapy products rely on dendritic cell-based systems or IL-2 expanded TIL. Hurdles remain in developing efficient, consistent, controlled processes; thus, these processes still have limited access to broad patient populations. Here, we describe a novel approach to produce cellular therapies at clinical scale, using proprietary nanoparticles combined with a proprietary manufacturing process to enrich and expand antigen-specific CD8<sup>+</sup> T-cell products with consistent purity, identity, and composition required for effective and durable anti-tumor response.


2020 ◽  
Vol 4 (19) ◽  
pp. 4653-4664
Author(s):  
Rajat K. Das ◽  
Roddy S. O’Connor ◽  
Stephan A. Grupp ◽  
David M. Barrett

Abstract Engineered T-cell therapies have demonstrated impressive clinical responses in patients with hematologic malignancies. Despite this efficacy, many patients have a transient persistence of T cells, which can be correlated with transient clinical response. Translational data on T cells from pediatric cancer patients shows a progressive decline in chimeric antigen receptor (CAR) suitability with cumulative chemotherapy regardless of regimen. We investigated the effects of chemotherapy on surviving T cells in vitro, describing residual deficits unique to each agent including mitochondrial damage and metabolic alterations. In the case of cyclophosphamide but not doxorubicin or cytarabine, these effects could be reversed with N-acetylcysteine. Specifically, we observed that surviving T cells could be stimulated, expanded, and transduced with CARs with preserved short-term cytolytic function but at far lower numbers and with residual metabolic deficits. These data have implications for understanding the effects of chemotherapy on mature T cells later collected for adoptive cell therapy, as chemotherapy-exposed T cells may have lingering dysfunction that affects ex vivo adoptive cell therapy manufacturing techniques and, ultimately, clinical efficacy.


2021 ◽  
Vol 12 ◽  
Author(s):  
Amrendra Kumar ◽  
Reese Watkins ◽  
Anna E. Vilgelm

The rationale behind cancer immunotherapy is based on the unequivocal demonstration that the immune system plays an important role in limiting cancer initiation and progression. Adoptive cell therapy (ACT) is a form of cancer immunotherapy that utilizes a patient’s own immune cells to find and eliminate tumor cells, however, donor immune cells can also be employed in some cases. Here, we focus on T lymphocyte (T cell)-based cancer immunotherapies that have gained significant attention after initial discoveries that graft-versus-tumor responses were mediated by T cells. Accumulating knowledge of T cell development and function coupled with advancements in genetics and data science has enabled the use of a patient’s own (autologous) T cells for ACT (TIL ACTs). In TIL ACT, tumor-infiltrating lymphocytes (TILs) are collected from resected tumor material, enhanced and expanded ex-vivo, and delivered back to the patient as therapeutic agents. ACT with TILs has been shown to cause objective tumor regression in several types of cancers including melanoma, cervical squamous cell carcinoma, and cholangiocarcinoma. In this review, we provide a brief history of TIL ACT and discuss the current state of TIL ACT clinical development in solid tumors. We also discuss the niche of TIL ACT in the current cancer therapy landscape and potential strategies for patient selection.


2013 ◽  
Vol 57 (1-3) ◽  
pp. 23-33 ◽  
Author(s):  
C. Marcela Díaz-Montero ◽  
Abdel-Aziz Zidan ◽  
Maria F. Pallin ◽  
Vasileios Anagnostopoulos ◽  
Mohamed L. Salem ◽  
...  

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.


Sign in / Sign up

Export Citation Format

Share Document