scholarly journals 855 Development of a novel anti-multiple myeloma chimeric antigen receptor T cell therapy

2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A896-A896
Author(s):  
Chuck Hay ◽  
Mary Faber ◽  
Kemi Adeyanju ◽  
Jeffrey Medin

BackgroundMultiple myeloma is a cancer of plasma cells, wherein the plasma cells begin outgrowing and even suppressing the growth of normal hematopoietic-lineage cells in the blood marrow. It is estimated that nearly 35,000 new cases of multiple myeloma will be diagnosed each year, and over 12,000 individuals will die from multiple myeloma in 2021. The current overall 5-year survival rate of 54% stresses the need for alternative treatment strategies. A chimeric antigen receptor T cell (CAR-T) therapy that targets multiple myeloma surface proteins may have the potential to improve this survival rate.MethodsA commonly associated multiple myeloma antigen was selected, and the extracellular domain of the protein was expressed in an Expi293 cell culture system. The multiple myeloma protein was expressed with and purified by a Hisx6 tag. The purified protein was used to pan a specific scFv phage display library that was generated uniquely for our lab from multiple blood donors. Several clones were identified and sequenced. One clone was selected and verified to be capable of binding the multiple myeloma antigen via ELISA. This anti-multiple myeloma scFv was then subcloned into CAR-T plasmids. This is a second-generation CAR-T using a 4-1BB co-stimulator domain and a CD3ζ intracellular signaling domain was used for this therapy. Further, a cell-fate control gene, LNGFRΔTmpk, was added to the plasmid to combat potential graft versus host disease. CAR-T and lentivirus (LV) plasmids were transfected into HEK cells to produce anti-multiple myeloma CAR-T LV.ResultsThe anti-multiple myeloma CAR LV was transduced into HEK cells and titered via qPCR, which showed the LV prep produced 3.1E8 IU/ml. qPCR also showed there to be 0.407 vector copies per cell on average when using an MOI of 3. Our CAR-T LV was then used to transduce Jurkat cells at an MOI of 3. Using anti-Fab and anti-protein L antibodies, the transduced cells were shown to be expressing the anti- multiple myeloma CAR-T by flow cytometry. After confirming successful transduction and expression, transduced Jurkats (and controls) were co-cultured with multiple myeloma cell lines and incubated for 24 hours. ELISA testing for IFN-γ was performed on the resulting supernatants; specific engagement was demonstrated.ConclusionsThese results show that our anti-multiple myeloma CAR is successfully expressed and is functional. Our future goal is to successfully transduce our anti-multiple myeloma CAR LV into primary T cells and test for functionality in these cells before transitioning into in vivo models.

2019 ◽  
Vol 12 (1) ◽  
Author(s):  
Chao Wu ◽  
Lina Zhang ◽  
Qierra R. Brockman ◽  
Fenghuang Zhan ◽  
Lijuan Chen

AbstractMultiple myeloma (MM) is the second most common hematologic malignancy and remains incurable despite the advent of numerous new drugs such as proteasome inhibitors (PIs), immunomodulatory agents (IMiDs), and monoclonal antibodies. There is an unmet need to develop novel therapies for refractory/relapsed MM. In the past few years, chimeric antigen receptor (CAR)-modified T cell therapy for MM has shown promising efficacy in preclinical and clinical studies. Furthermore, the toxicities of CAR-T cell therapy are manageable. This article summarizes recent developments of CAR-T therapy in MM, focusing on promising targets, new technologies, and new research areas. Additionally, a comprehensive overview of antigen selection is presented along with preliminary results and future directions of CAR-T therapy development.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Faroogh Marofi ◽  
Safa Tahmasebi ◽  
Heshu Sulaiman Rahman ◽  
Denis Kaigorodov ◽  
Alexander Markov ◽  
...  

AbstractDespite many recent advances on cancer novel therapies, researchers have yet a long way to cure cancer. They have to deal with tough challenges before they can reach success. Nonetheless, it seems that recently developed immunotherapy-based therapy approaches such as adoptive cell transfer (ACT) have emerged as a promising therapeutic strategy against various kinds of tumors even the cancers in the blood (liquid cancers). The hematological (liquid) cancers are hard to be targeted by usual cancer therapies, for they do not form localized solid tumors. Until recently, two types of ACTs have been developed and introduced; tumor-infiltrating lymphocytes (TILs) and chimeric antigen receptor (CAR)-T cells which the latter is the subject of our discussion. It is interesting about engineered CAR-T cells that they are genetically endowed with unique cancer-specific characteristics, so they can use the potency of the host immune system to fight against either solid or liquid cancers. Multiple myeloma (MM) or simply referred to as myeloma is a type of hematological malignancy that affects the plasma cells. The cancerous plasma cells produce immunoglobulins (antibodies) uncontrollably which consequently damage the tissues and organs and break the immune system function. Although the last few years have seen significant progressions in the treatment of MM, still a complete remission remains unconvincing. MM is a medically challenging and stubborn disease with a disappointingly low rate of survival rate. When comparing the three most occurring blood cancers (i.e., lymphoma, leukemia, and myeloma), myeloma has the lowest 5-year survival rate (around 40%). A low survival rate indicates a high mortality rate with difficulty in treatment. Therefore, novel CAR-T cell-based therapies or combination therapies along with CAT-T cells may bring new hope for multiple myeloma patients. CAR-T cell therapy has a high potential to improve the remission success rate in patients with MM. To date, many preclinical and clinical trial studies have been conducted to investigate the ability and capacity of CAR T cells in targeting the antigens on myeloma cells. Despite the problems and obstacles, CAR-T cell experiments in MM patients revealed a robust therapeutic potential. However, several factors might be considered during CAR-T cell therapy for better response and reduced side effects. Also, incorporating the CAT-T cell method into a combinational treatment schedule may be a promising approach. In this paper, with a greater emphasis on CAR-T cell application in the treatment of MM, we will discuss and introduce CAR-T cell’s history and functions, their limitations, and the solutions to defeat the limitations and different types of modifications on CAR-T cells.


2017 ◽  
Vol 35 (18_suppl) ◽  
pp. LBA3001-LBA3001 ◽  
Author(s):  
Frank (Xiaohu) Fan ◽  
Wanhong Zhao ◽  
Jie Liu ◽  
Aili He ◽  
Yinxia Chen ◽  
...  

LBA3001 Background: Chimeric antigen receptor engineered T cell (CAR-T) is a novel immunotherapeutic approach for cancer treatment and has been clinically validated in the treatment of acute lymphoblastic leukemia (ALL). Here we report an encouraging breakthrough of treating multiple myeloma (MM) using a CAR-T designated LCAR-B38M CAR-T, which targets principally BCMA. Methods: A single arm clinical trial was conducted to assess safety and efficacy of this approach. A total of 19 patients with refractory/relapsed multiple myeloma were included in the trial. The median number of infused cells was 4.7 (0.6 ~ 7.0) × 10e6/ kg. The median follow-up times was 208 (62 ~ 321) days. Results: Among the 19 patients who completed the infusion, 7 patients were monitored for a period of more than 6 months. Six out of the 7 achieved complete remission (CR) and minimal residual disease (MRD)-negative status. The 12 patients who were followed up for less than 6 months met near CR criteria of modified EBMT criteria for various degrees of positive immunofixation. All these effects were observed with a progressive decrease of M-protein and thus expected to eventually meet CR criteria. In the most recent follow-up examination, all 18 survived patients were determined to be free of myeloma-related biochemical and hematologic abnormalities. One of the most common adverse event of CAR-T therapy is acute cytokine release syndrome (CRS). This was observed in 14 (74%) patients who received treatment. Among these 14 patients there were 9 cases of grade 1, 2 cases of grade 2, 1 case of grade 3, and 1 case of grade 4 patient who recovered after treatments. Conclusions: A 100% objective response rate (ORR) to LCAR-B38M CAR-T cells was observed in refractory/relapsed myeloma patients. 18 out of 19 (95%) patients reached CR or near CR status without a single event of relapse in a median follow-up of 6 months. The majority (14) of the patients experienced mild or manageable CRS, and the rest (5) were even free of diagnosable CRS. Based on the encouraging safety and efficacy outcomes, we believe that our LCAR-B38M CAR-T cell therapy is an innovative and highly effective treatment for multiple myeloma.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 381-381 ◽  
Author(s):  
Bijan Boldajipour ◽  
Roman Galetto ◽  
Cesar Sommer ◽  
Thomas Pertel ◽  
Julien Valton ◽  
...  

Abstract Multiple myeloma (MM) is a hematological disease of plasma B cells that remains incurable despite the availability of numerous therapies. The plasma cell-specific expression of the TNF superfamily receptor BCMA may allow targeting of normal and malignant plasma cells. Genetically engineered chimeric antigen-receptor T cells (CAR T) have shown tremendous promise in the treatment of several hematological diseases, including MM. However, conventional autologous CAR T therapies use patient-derived T cells and the logistics of on-demand CAR T manufacture limits their availability to a broad patient pool. Here we describe the preclinical evaluation of an allogeneic CAR T therapy targeting BCMA that has the potential for a readily available, off-the-shelf therapy for MM and other malignancies expressing BCMA. Human T cells were transduced with recombinant lentiviral vectors encoding three BCMA CAR candidates designed with fully human anti-BCMA scFvs, CD8a transmembrane domains and the intracellular signaling domains of 4-1BB and CD3zeta. All CAR T efficiently killed BCMA-expressing multiple myeloma cell lines (KMS12BM, MM1.S, Molp-8 and OPM-2), but not BCMA-negative REH cells in vitro and in vivo. Whereas 2 of the 3 candidates exhibited target-independent cytokine production, accelerated T cell differentiation and reduced target cell-induced expansion in vitro, the third candidate did not exhibit this scFv-induced autoactivation and was chosen as the lead molecule. Due to the allogeneic nature of this T cell therapy, the possibility of graft-versus-host (GvH) reactions can be a safety concern. We applied Cellectis' know-how and TALEN® technology for the gene inactivation of the T cell receptor (TCR) alpha chain to significantly reduce the probability for TCR-mediated GvH reactions and found that TCR knockout did not affect CAR T activity in vitro or in vivo. Furthermore, we incorporated intra-CAR rituximab-recognition domains into the CAR molecule to enable depletion of CAR T cells from patients when necessary. We found that this modified CAR retained anti-BCMA CAR T activity and enabled CAR T depletion by rituximab. Another aspect of allogeneic CAR T therapies is the rejection of the CAR T by host-versus-graft (HvG) reactions. Lymphodepletion prior to CAR T infusion enhances CAR T efficacy in autologous CAR T trials and may also prevent anti-CAR HvG reactions in allogeneic therapy settings. Engineering lymphodepletion resistance into CAR T cells could therefore enable sustained lymphodepletion for enhanced allogeneic CAR T persistence and efficacy. CD52 is expressed on all lymphocytes and administration of the anti-CD52 antibody alemtuzumab for prolonged lymphodepletion is an approved treatment for multiple sclerosis. TALEN®-mediated knockout of CD52 protected BCMA CAR T from alemtuzumab-induced cytotoxicity and did not alter BCMA CAR T anti-tumor activity. Taken together these results support allogeneic BCMA CAR T as an off-the-shelf adoptive immunotherapy for the treatment of multiple myeloma and other BCMA-positive malignancies. Disclosures Boldajipour: Pfizer: Employment. Galetto:Cellectis SA: Employment. Sommer:Pfizer Inc.: Employment. Pertel:Pfizer Inc.: Employment. Valton:Cellectis Inc.: Employment. Park:Pfizer Inc.: Employment. Gariboldi:Cellectis SA: Employment. Chen:Alexo Therapeutics: Employment. Geng:Kodiak Sciences: Employment. Dong:Pfizer Inc.: Employment. Boucher:Pfizer Inc.: Employment. Van Blarcom:Pfizer Inc.: Employment. Chaparro-Riggers:Pfizer Inc.: Employment. Rajpal:Pfizer Inc.: Employment. Smith:Cellectis SA: Employment. Kuo:Pfizer Inc.: Employment. Sasu:Pfizer Inc.: Employment.


Sign in / Sign up

Export Citation Format

Share Document