Progress and prospects for use of cellular immunotherapy in pancreatic cancer


 Table of Contents   REVIEW ARTICLE Year : 2022  |  Volume : 18  |  Issue : 7  |  Page : 1867-1875

Progress and prospects for use of cellular immunotherapy in pancreatic cancer

Jing Tian1, Tiankai Bai1, Zhiyong Zhang2, Xuan Zhai1, Kangmin Wang1, Xingyi Gao1, Bin Yan1
1 School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
2 School of Basic Medicine, Shandong First Medical University, Jinan, China

Date of Submission17-Jun-2021Date of Decision22-Mar-2022Date of Acceptance29-Oct-2022Date of Web Publication11-Jan-2023

Correspondence Address:
Bin Yan
School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Daxue Road 4655, Changqing District, Jinan - 250355
China
Login to access the Email id

Source of Support: None, Conflict of Interest: None

Crossref citationsCheck

DOI: 10.4103/jcrt.jcrt_976_21

Rights and Permissions


Pancreatic cancer (PC) is a highly malignant tumor with an increasing incidence rate in recent years. Because pancreatic cancer has an insidious onset, unknown pathophysiology, and poor prognosis, the overall survival rate of pancreatic cancer patients has not improved considerably even with extensive treatment methods such as surgery, radiation, biotherapy, and targeted therapy. Therefore, finding and developing more effective and safe treatments for pancreatic cancer is critical. Cellular immunotherapy has achieved considerable advances in the field of oncology in recent years. Technology is continuously advancing, with new breakthroughs virtually every month, and pancreatic cancer eradication is expected to improve considerably. This article examines the advance of chimeric antigen receptor NK cell immunotherapy (CAR-NK) cell immunotherapy for pancreatic cancer research, as well as research ideas for pancreatic cancer treatment.

Keywords: CAR-NK cell immunotherapy, CAR-T cell immunotherapy, pancreatic cancer, research progress


How to cite this article:
Tian J, Bai T, Zhang Z, Zhai X, Wang K, Gao X, Yan B. Progress and prospects for use of cellular immunotherapy in pancreatic cancer. J Can Res Ther 2022;18:1867-75
How to cite this URL:
Tian J, Bai T, Zhang Z, Zhai X, Wang K, Gao X, Yan B. Progress and prospects for use of cellular immunotherapy in pancreatic cancer. J Can Res Ther [serial online] 2022 [cited 2023 Jan 13];18:1867-75. Available from: https://www.cancerjournal.net/text.asp?2022/18/7/1867/367482  > Introduction Top

Pancreatic cancer (PC) is a cancerous tumor that affects the pancreas. Because the pathophysiology of pancreatic cancer is unknown, early detection and treatment of the disease are extremely difficult. As a result, the number of patients diagnosed with pancreatic cancer is rising over the world.[1],[2],[3],[4] According to the Global Cancer Data 2020, pancreatic cancer affects about 495,773 people worldwide in one year, with more than 46,000 individuals dying from the disease. Pancreatic cancer patients have a 5-year survival rate of fewer than 10 years.[5] Surgery is currently the main treatment approach for pancreatic cancer in its early stages. Palliative short-circuit surgery,[6] chemotherapy, and radiotherapy[7] are commonly used to treat inoperable patients, although many individuals have developed resistance to these treatments.[8] Pancreatic cancer is still one of the most lethal types of cancer.[9] Therefore, more effective treatments for pancreatic cancer are required.

Cellular immunotherapy has made substantial advances in the treatment of tumors in recent years.[10] It is a technique that involves collecting human autoimmune cells, genetically modifying and culturing them in vitro to boost specific killing activity, and then returning them to the human body to kill pathogens, cancer cells, and mutant cells in blood and tissues. For example, chimeric antigen receptor T cell immunotherapy (CAR-T) has demonstrated significant benefits in the treatment of hematologic cancers while overcoming many of the limitations of conventional therapies.[11],[12] Six CAR-T therapy medicines have been approved for marketing by the U.S. FDA according to incomplete statistics, ushering in a new era in cancer treatment. CAR-T immunotherapy is projected to enhance cancer patient survival rates when treating cancer. Although CAR-T cell immunotherapy has made significant progress in recent years, its severe neurotoxicity and cytokine storm have limited its use in clinics. Additional cell-mediated cytotoxic immunotherapies are urgently needed to overcome the disadvantages of CAR-T cell immunotherapy.

NK cells are a safe and effective alternative immunotherapy technique for T cells in clinical practice due to their distinct biological properties. Genetic engineering is also used in NK cell immunotherapy to add chimeric antibodies (CARs) to NK cells, transforming them into CAR-NK cells that detect tumor cells while activating NK cells to kill tumor cells, boosting NK cells' ability to target and kill tumor cells. Immunotherapy using CAR-T and CAR-NK cells is a promising new advance in the treatment of solid malignancies. As a result, this article provides a brief overview of the research progress of CAR-T cells and CAR-NK cells for pancreatic cancer treatment, including the basic composition of CAR-T cells and CAR-NK cells, the currently popular pancreatic cancer targets, and the major challenges and solutions they face, as well as research ideas for pancreatic cancer treatment.

 > The Basic Composition of Chimeric Antigen Receptors (CARS) Top

A tumor-associated antigen (TAA) binding region (usually derived from the ScFv segment of the monoclonal antibody-antigen binding region), an extracellular hinge region, a transmembrane region, and an intracellular signal transduction region made up the basic design of Chimeric antigen receptors (CARs). When ScFv fragments connect to tumor antigens selectively, they activate intracellular signaling regions in CAR-T or CAR-NK cells, causing them to activate, proliferate and kill tumor cells. This enables CAR-T cells or CAR-NK cells to recognize a greater number of targets than naive T cells or NK cells.[13]

 > Popular Targets for Pancreatic Cancer Top

The choice of target antigens for Chimeric antigen receptor immune cell treatment for solid tumors is important to success.[14] For the therapy of solid tumors, several target antigens have been identified. For example, a 7-year-old boy in Texas, USA, who was diagnosed with myeloid metastatic rhabdomyosarcoma and treated with HER2-CAR-T cells immunotherapy in July 2020 has been cancer-free for 19 months and no cancer cells have been detected in his body.[15] In 2020, a patient with advanced ovarian cancer who received PD-1-MESO-CAR-T cells paired with apatinib treatment at Shanghai People's Hospital, China, was in remission and lived for more than 17 months.[16] Professor Wendell Lim led a research team that produced synNotch-CAR-T, a new form of CAR-T cell that is effective against solid tumors and can completely eradicate glioblastomas of human origin in mice's brains[17]

Similarly, pancreatic cancer cells have a variety of tumor-associated antigen epitopes on their surface [Table 1] and [Table 2], which gives CAR-T or CAR-NK immunotherapy for pancreatic cancer treatment more target options.

 > Challenges of CAR-T Cell Immunotherapy for Pancreatic Cancer and Strategies to Address Them Top

CAR-T cell immunotherapy, a strong tumor-killing cross-generational treatment, is still being studied for its efficacy and safety. CAR-T immunotherapy has numerous hurdles in the treatment of pancreatic cancer[35]: (1) Tumor microenvironment suppression. (2) Cytokine release syndrome is a condition in which cytokines are released into the body. (3) Failure of CAR-T cells. Furthermore, the majority of antigens discovered in solid tumors are tumor-associated antigens that can be expressed in other normal tissues, creating the possibility of “antigen escape” during CAR-T cell immunotherapy.

Suppression of tumor microenvironment and strategies to address it

In the stroma of solid tumors, many tumor-associated fibroblasts secrete collagen and produce dense tumor tissue, resulting in abnormally increased interstitial pressure and a physical barrier to CAR-T cell infiltration.[36] As a result, boosting CAR-T cell infiltration to reverse immune microenvironment suppression should improve the efficacy of CAR-T immunotherapy in solid tumors.[37],[38] The use of cutting-edge nanotechnology has considerably enhanced CAR-T cell targeting and increased CAR-T cell infiltration. The use of adjuvant-loaded lipid nanoparticles (LNP) in combination with medications in cancer immunotherapy has been shown to improve drug response to tumors, according to Nakamura T.[39] Drugs can reach the lesion site and have a therapeutic impact by breaking through the suppression of the tumor microenvironment using lipid nanoparticles (LNP) loaded with adjuvants. As a photosensitizer and drug carrier, Hao Y[40] created a near-infrared sensitive inorganic CuS nanoparticle. Due to the presence of folic acid on its surface, the CuS delivers the medicine into the tumor cells when injected intratumorally. The nanocarriers disintegrate under laser irradiation and release therapeutic medicines, which can be used in conjunction with photothermal therapy to ablate tumor cells and increase immunogenic cell death. These smart nanocarriers may also prevent CAR-T cells from entering tumors, allowing them to unleash their potent anti-tumor immune response and provide long-term immunological protection against tumor recurrence. Xia H[41] describes a TLR7/8 agonist-conjugated nanovaccine (TNV) that intelligently responds to the acidic environment and endosomal cathepsin precisely releases TLR7/8 agonist to activate receptor signaling at the endosomal membrane, stimulates DCs maturation, and provokes specific cellular immunity. TNV's preventive and therapeutic efficacy in mouse melanoma and colon cancer has been demonstrated in vivo. Miller IC[42] and his colleagues also introduced a genetic switch to CAR-T cells, allowing them to transport genetically engineered T cells to the tumor microenvironment precisely. After that, laser pulses were used to boost the temperature of the mouse tumor to 40–42°C, which activated the CAR-T cell gene switch and increased the expression of anti-cancer proteins. This overcomes the tumor microenvironment's inhibition and prevents tumor recurrence. To achieve rapid intracellular substance delivery and subsequent reversal of cell permeability, Kavanag HH's team[43] utilized reversible osmosis technology. In terms of manufacturing, safety, and regulation, it has potential advantages over viral vectors. Although electroporation is the most extensively used non-viral form of cell engineering, it can cause damage to cells. Lysis, on the other hand, is the least harmful to human T cells. Castellari M[44],[45] developed a switchable immune receptor Sortases CAR (Srt.bbz), which can connect to several tumor antigens and allow CAR-T cells to switch to recognize their target antigens, and preliminary testing has demonstrated that Srt.bbz T cells can lyse tumor cells.

In addition, Y-mAbs has developed naxitamab, a humanized monoclonal antibody that binds to GD2 antigen on the tumor surface and activates the complement system in the immune system, triggering an antibody-mediated cytotoxic response and activating the complement system in the immune system, preclinical data shows that it aids CAR-T reversing immunosuppression of the cellular tumor microenvironment. Researchers at City of Hope in the United States have developed a new form of B cell activating factor receptor (BAFF-R) CAR-T cells that target the CD19 antigen and has a better ability to bind to tumor cells by replacing the CD19 target with the B cell activating factor receptor (BAFF-R). The development of these technologies enhances the milieu for CAR-T therapy in the treatment of malignancies, and it may also improve pancreatic cancer treatment.

Cytokine release syndrome (CRS) and strategies to address it

The excessive release of cytokines including IL-6 and IL-10 at the moment of T cell activation following CAR-T treatment causes cytokine release syndrome (CRS).[46],[47] CAR-T cell treatments that target CD19, BCMA, and CD22 have all resulted in severe cytokine crises so far.[48] This necessitates inserting a “control gene” into the CAR-T gene, which effectively prevents cytokine storms. It's still a big problem to avoid cytokine storms after CAR-T treatment while pancreatic cancer treatment is still in preclinical or early clinical trials.

CAR-T cell failure and strategies to address it

CAR-T has achieved several advancements in hematological cancers, but solid tumors have long been a barrier. The main cause of CAR-T failure is non-antigen-dependent activation, which has become the main optimization route for CAR-T cells to combat pancreatic cancer. Some researchers, for example, have developed the STAR double-chain chimeric receptor, which integrates the antigen recognition and TCR activation pathways of CAR-T [Figure 1] and has higher antigen sensitivity and improved T cell persistence than CAR-T cells, making it a promising treatment for pancreatic cancer.[49] The Weberew S team[50] discovered that epigenetic modification can restore the function of failing CAR-T cells. Proline dehydrogenase 2 (PRODH2) has also been found to improve the therapeutic efficacy of CAR-T cells in a variety of animal cancer models.[51] They used a dead-guide RNA (dgRNA)-based CRISPR activation screen in primary CD8+ T cells to find a CAR-T engineering gain-of-function target. PRODH2 engineering improved the antitumor metabolism and immunological function of CAR-T cells, and PRODH2 was a target for improving CAR-T cell efficacy, according to the researchers.

Antigen escape and strategies to address it

One of the most successful approaches to address antigen escape is to investigate pancreatic cancer tumor surface-specific antigens. However, all of the antigens discovered so far are tumor-associated antigens. As a result, researchers expect to lessen the “off-target” effect by developing dual-target CAR-T cells. In a phase, I trial in patients with relapsed/refractory pre-acute B-lymphoblastic leukemia (B-ALL), Dai H[52]et al. demonstrated that bispecific CD19/CD22 CAR-T cells can have strong cytolytic activity against target cells and induce relapsed/refractory B-ALL in adult patients with a remission effect. Antigen escape can also be caused by low antigen expression in solid tumors, and how to tackle this problem will be a future focus of CAR-T therapy.

 > Challenges of CAR-NK Cell Immunotherapy for Pancreatic Cancer Top

CAR-NK cells offer distinct advantages over CAR-T cells, although they still face significant obstacles. Improvements in NK cell proliferation, activation of NK cell toxicity, and the best technique to rebuild NK cells are among the issues.

In vitro expansion of NK cells is the first hurdle for CAR-NK cell immunotherapy

Because a single donor's NK cells are insufficient for treatment, it is critical to figure out how to multiply NK cells in vast quantities. Although it has been discovered that irradiation improves NK cell multiplication, the lack of donor cell numbers remains a hurdle.[53] To avoid the development of GVHD (graft-versus-host disease), T cells must be totally eliminated when treating tumors with CAR-NK cells. As a result, getting enough NK cells remains a challenge.

CAR-NK cell construction is the second hurdle for CAR-NK cell immunotherapy

Retroviruses and lentiviruses have been employed to transfect CARs in the past. Although retroviral vectors have a high transfection efficiency, they can produce insertional mutations, cancer, and other side effects. Using lentiviral vectors, on the other hand, despite the low occurrence of insertional mutations, the efficacy of transfecting NK cells was similarly reduced. As a result, increasing the efficiency of CAR transfection provides another avenue for tumor therapy.

 > Innovative Combination Therapy Top

Immune cell treatments' therapeutic potential can be boosted by combining immunosuppressive medications, targeted medicines, or other techniques, in addition to optimizing CARs themselves.

Combined therapy with CRISPR/Cas9 gene technology

CRISPR-associated nuclease9 (CRISPR/Cas9) technology, a bacterial and archaeal-acquired immune system, has provided a new tool for precise genome editing. It was a simple and flexible genome editing method that targeted practically any genomic locus by using only a single nuclease protein in conjunction with two short RNA as a site-specific endonuclease. CRISPR/Cas9 technology as a genetic editing tool in combination with CAR-T cell treatment has been shown in studies to improve the efficiency and safety of CAR-T cells. CRISPR/Cas9 technology was used by Zhang S[54] to knock down the GPR54 or ERK5 gene in CAR-T cells, which improved the anti-tumor response of CAR-T cells to PSMA+ and CD19+ tumor cells while avoiding T cells depletion. Alishah K[55] used the CRISPR/Cas9 system to knock out the TGFβRII gene in T cells, allowing CAR-T cells to proliferate and inhibit tumors more effectively. CRISPR/Cas9 technology has also been used in CAR-NK cell therapy to improve NK cell anti-tumor function. CRISPR/Cas9 technology was employed by May Daher et colleagues[56] to remove the CISH gene in CAR-NK cells, which improved their metabolic capacity and anti-tumor effectiveness. CRISPR/Cas9 technology was utilized by one team[57] to disrupt the CD38 gene during CAR-NK cells expansion and knocking down the CD38 gene NK cells reduced cell self-mutilation and improved the treatment of acute myeloid leukemia (AML). All of these findings point to CRISPR/Cas9 gene editing technology being able to improve the efficacy of cellular immunotherapy in cancer treatment.

Combined therapy with immune checkpoint inhibitors

Scholars focused solely on improving T-cell activation in the early days of pancreatic cancer treatment. Researchers didn't know that T-cell activation was followed by activation of the suppressor t-cell pathway until the late 1980s. Immune checkpoint inhibitors, such as monoclonal antibody inhibitors, stimulate systemic immunity and boost T-cell activity by blocking T-cell inhibitory signaling pathways. Immune checkpoint inhibitors can thereby slow tumor progression and possibly eliminate tumors.[58],[59],[60],[61],[62],[63],[64],[65],[66],[67] Similarly, checkpoint inhibitors can enhance CAR-T cells' proliferative potential by blocking the inhibitory signaling pathway. As a result, they work together to prevent antigen escape and improve the targeting and tumor-killing abilities of CAR-T cells. To treat pancreatic cancer, Ching Y[68] employed chimeric antigen receptor T cells in combination with immune checkpoint inhibitors PD-1/PD-L1. In vitro tests revealed that 80 percent of tumor cells overexpressing PD-L1 could be detected and eliminated [Figure 2]. Chong EA[69] also described a case of a patient with refractory diffuse B large cell lymphoma who was unresponsive after therapy with CD19-CAR-T cells, but whose symptoms were reduced after co-treatment with pembrolizumab, which increased CAR-T cells activity and improved CAR-T cells activity. CAR-T cells paired with immune checkpoint inhibitors may be successful in treating solid tumors, according to the American Cancer Society in 2019. Grosser[70] further stated that using immune checkpoint inhibitors alone to treat pancreatic cancer is currently ineffective. These findings give a theoretical foundation for using CAR-T cells in combination with checkpoint inhibitors. In tumor patients, NK cells have been found to exhibit the inhibitory checkpoint PD-1, according to recent research. As a result, checkpoint inhibitors may improve the effectiveness of CAR-NK cells against PD-L1-positive malignancies.[71]

Combined therapy with oncolytic viruses

Oncolytic viruses (OVs) selectively infect tumor cells and replicate in large numbers, eventually lysing the cells and expanding the invasion space of immune cells in tumor tissues,[72],[73] suggesting that OVs combined with CAR-T or CAR-NK therapy could break through the immune microenvironment of pancreatic cancer.[72],[73] Transgenic OVs can release cellular immune components, activate the body's immune system, and target and kill tumor cells after penetrating tumor cells. Reoviridae, HSV virus, and tumor lysing adenovirus can all be utilized as vectors to deliver encoded gene snippets to tumor cells. They can lyse the extracellular matrix of pancreatic cancer cells in this way, improving the immunological microenvironment of pancreatic cancer, increasing tumor cell exposure, and inhibiting pancreatic cancer metastasis. OVs have a wide host range, similarity with human genes, and no mutation when introduced into the host, therefore it may be employed as a viral vector to create CAR-T or CAR-NK cells, compared to other viral vectors. Following that, a group of researchers discovered that OV19s and CD19-CAR-T cells had a remarkable synergistic impact, killing cancer cells in large numbers. In a glioblastoma (GBM) mouse model, Ma R[74] discovered that combining OVs expressing IL15/IL15Rα complexes with EGFR-CAR-NK cells increased overall survival and dramatically suppressed tumor development compared to monotherapy. As a result, using mildly pathogenic viruses could improve the efficiency of cellular immunotherapy for pancreatic cancer while also providing new research ideas.

Combined therapy with targeted drugs

Many molecularly targeted therapeutic drugs have first shown anti-pancreatic cancer properties, thanks to the rapid development of contemporary immunology and molecular biology. Epidermal growth factor (EGFR) receptor-targeting medications, vascular endothelial growth factor targeting drugs, anti-angiogenic matrix metalloproteinase inhibitors (MMPIs), and other pharmaceuticals are commonly used. In pancreatic cancer treatment, cetuximab inhibits EGFR phosphorylation, decreases VEGF production, and prevents neovascularization, increasing the toxicity of CAR-T or CAR-NK cells.

Combined therapy with antineoplastic drugs

By modulating cytokines or down-regulating the PD-1/PDL1 pathway, some standard anti-tumor medications [Table 3] can suppress tumor cell growth and migration. When these medications are used with cellular immunotherapy, immune cells are more likely to connect to specific antigens or related antigens on the surface of tumor cells, resulting in a synergistic effect with immune cells in the treatment of cancer.[75],[76]

 > Conclusion Top

Together, with the advancement of modern technology and the efforts of researchers from numerous countries, there is a great deal of optimism that cancer can be defeated. At the moment, it's difficult to entirely eradicate malignancies with a single treatment. Combination immunotherapy for cancers will be the main research focus in the future, although traditional methods will continue to play an essential part in the treatment process. Genetic engineering, which can repair and modify mutations or gene deletions and is a milestone in the radical cure or prevention of cancer, has many advantages in addition to immunotherapy. This strategy, however, still has a number of flaws. Although gene therapy is theoretically conceivable, few gene therapy approaches have been used in clinical practice, and the majority of them are still in the research and animal testing stages. In the United States, a patient died after receiving gene therapy.

The medicine has major repercussions since it entered the body without replacing or deleting the disease-causing gene. Furthermore, numerous teams have made substantial advances in the use of TCR-T for cancer treatment, implying that TCR-T can identify a broader spectrum of potential tumor-specific antigens or improve tumor-killing effects.[82],[83] Clinical trials using TCR-T to treat various types of cancer are now being conducted by companies such as Adaptimmune, Immunocore and TCR2 Therapeutics. With the success of CAR-T cell therapy and the huge potential of CAR-NK cell therapy, scientists are looking into the ability of mononuclear macrophages (MPS) to phagocytose exogenous microorganisms once more. CAR-Macrophages (CAR- M) have been shown to have considerable benefits in the treatment of solid malignancies in preliminary investigations. It solves the problem of CAR-T cells invading the tumor microenvironment and offers a lot of research potential. The FDA has approved two clinical trials based on the CAR-M approach, providing yet another research avenue for pancreatic cancer treatment. The eradication of solid tumors is seen to be inevitable as technology improves.

Financial support and sponsorship

Shandong Provincial Medical and Health Science and Technology Development Program (No.:202002050626), the Subproject of Major New Drug Innovation and Technology Special Project of the Ministry of Health of China (2014ZX09509001001).

Conflicts of interest

There are no conflicts of interest.

 

 > References Top
1.Frappart PO, Hofmann TG. Pancreatic ductal adenocarcinoma (PDAC) organoids: The shining light at the end of the tunnel for drug response prediction and personalized medicine. Cancers (Basel) 2020;10:2750.  Back to cited text no. 1
    2.Park TH, Kim HS. Eupatilin suppresses pancreatic cancer cells via glucose uptake inhibition, AMPK activation, and cell cycle arrest. Anticancer Res 2022;1:483-91.  Back to cited text no. 2
    3.Hernández-Garduño E. The association between diabetes and cancer in Mexico: Analysis using death certificate databases, 2009-2017. J Cancer Res Ther 2021;6:1397-403.  Back to cited text no. 3
    4.Gong W, Zou Y, Wang P, Wang H. Relationship between systemic inflammatory response index and fibrinogen level with chemotherapy efficacy and prognosis in patients with advanced pancreatic cancer. J Chin Pract Diagn Ther 2021;12:1224-28.  Back to cited text no. 4
    5.Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: Globo estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021;3:209-49.  Back to cited text no. 5
    6.Aoyama T, Kazama K, Murakawa M, Atsumi Y, Shiozawa M, Ueno M, et al. Safety and feasibility of enhanced recovery after surgery in the patients underwent distal pancreatectomy for pancreatic cancer. J Cancer Res Ther 2018;14:S724-9.  Back to cited text no. 6
    7.Gai B, Li Q, Shao P, Yang D. Advantages of interstitial radioactive seed implantation for the treatment of Stage III pancreatic cancer. J Cancer Res Ther 2021;3:702-6.  Back to cited text no. 7
    8.Yimo Y, Xiaodong T. Guidelines for the diagnosis and treatment of pancreatic cancer in China. Chin J Pract surg 2021;7:725-38.  Back to cited text no. 8
    9.Fan X. Comparison of the efficacy and prognosis of radiotherapy with different segmentation Modalities in patients with inoperable resectable pancreatic[D]. Jilin University 2021.  Back to cited text no. 9
    10.Yeo D, Giardina C, Saxena P, Rasko JEJ. The next wave of cellular immunotherapies in pancreatic cancer. Mol Ther Oncolytics 2022;24:561-76.  Back to cited text no. 10
    11.Fang J, Ding N, Guo X, Sun Y, Zhang Z, Xie B, et al. αPD-1-mesoCAR-T cells partially inhibit the growth of advanced/refractory ovarian cancer in a patient along with daily apatinib. Immunother Cancer 2021;2:e1162.  Back to cited text no. 11
    12.Choe JH, Watchmaker PB, Simic MS, Gilbert RD, Li AW, Krasnow NA, et al. SynNotch-CAR -T cells overcome challenges of specificity, heterogeneity, and persistence in treating glioblastoma. Transl Med 2021;591:eabe7378.  Back to cited text no. 12
    13.Zhang C, Liu J, Zhong JF, Zhang X. Engineering CAR-T cells. Biomark Res 2017;5:22.  Back to cited text no. 13
    14.Leko V, Rosenberg SA. Identifying and targeting human tumor antigens for T cell-based immunotherapy of solid tumors. Cancer Cell 2020;38:454-72.  Back to cited text no. 14
    15.Hegde M, Joseph SK, Pashankar F, DeRenzo C, Sanber K, Navai S, et al. Tumor response and endogenous immune reactivity after administration of HER2 CAR-T cells in a child with metastatic rhabdomyosarcoma. Nat Commun 2020;11:3549.  Back to cited text no. 15
    16.Lee HH, Kim I, Kim UK, Choi SS, Kim TY, Lee D, et al. Therapeutic effiacy of T cells expressing chimeric antigen receptor derived from a mesothelin-specific scFv in orthotopic human pancreatic cancer animal models. Neoplasia 2022;2:98-108.  Back to cited text no. 16
    17.Hyrenius-Wittsten A, Su Y, Park M, Garcia JM, Alavi J, Perry N, et al. SynNotch CAR circuits enhance solid tumor recognition and promote persistent antitumor activity in mouse models. Transl Med2021;591:eabd8836.  Back to cited text no. 17
    18.Liang J, Zhang H, Huang Y, Fan L, Li F, Li M, et al. A CLDN18.2-targeting bispecific T cell co-stimulatory activator for cancer immunotherapy. Cancer Manag Res 2021;13:6977-87.  Back to cited text no. 18
    19.Ko AH, Jordan AC, Tooker E, Lacey SF, Chang RB, Li Y, et al. Dual targeting of mesothelin and CD19 with chimeric antigen receptor-modified T Cells in patients with metastatic pancreatic cancer. Mol Ther 2020;28:2367-78.  Back to cited text no. 19
    20.Pang N, Shi J, Qin L, Chen A, Tang Y, Yang H, et al. IL-7 and CCL19-secreting CAR-T cell therapy for tumors with positive glypican-3 or mesothelin. J Hematol Oncol 2021;1:118.  Back to cited text no. 20
    21.He J, Zhang Z, Lv S, Liu X, Cui L, Jiang D, et al. Engineered CAR-T cells targeting mesothelin by piggyBac transposon system for the treatment of pancreatic cancer. Cell Immunol 2018;329:31-40.  Back to cited text no. 21
    22.Golubovskaya V, Berahovich R, Zhou H, Xu S, Harto H, Li L, et al. CD47-CAR-T cells effectively kill target cancer cells and block pancreatictumor growth. Cancers (Basel) 2017;10:139.  Back to cited text no. 22
    23.Liu Y, Guo Y, Wu Z, Feng K, Tong C, Wang Y, et al. Anti-EGFR chimeric antigen receptor-modified T cells in metastatic pancreatic carcinoma: A phase I clinical trial. Cytotherapy 2020;22:573-80.  Back to cited text no. 23
    24.Zhang E, Yang P, Gu J, Wu H, Chi X, Liu C, et al. Recombination of a dual-CAR-modified T lymphocyte to accurately eliminate pancreatic malignancy. J Hematol Oncol 2018;11:102.  Back to cited text no. 24
    25.Raj D, Nikolaidi M, Garces I, Lorizio D, Castro NM, Caiafa SG, et al. CEACAM7 is an effective target for CAR T-cell therapy of pancreatic ductal adenocarcinoma. Clin Cancer Res 2021;27:1538-52.  Back to cited text no. 25
    26.Posey AD Jr, Schwab RD, Boesteanu AC, Steentoft C, Mandel U, Engels B, et al. Engineered CAR-T cells targeting the cancer-associated Tn-Glycoform of the membrane mucin MUC1 control adenocarcinoma. Immunity 2016,44:1444-54.  Back to cited text no. 26
    27.Reader CS, Vallath S, Steele CW, Haider S, Brentnall A, Desai A, et al. The integrin αvβ6 drives pancreatic cancer through diverse mechanisms and represents an effective target for therapy. J Pathol 2019;3:332-42.  Back to cited text no. 27
    28.Cong P, Yi C, Wang X, Peng Y. Construction of specific Smo lentivirus and expression of infected pancreatic cancer cells positive for CD24, CD44 surface antibody. J Biol Regul Homeost Agents 2021;2:525-35.  Back to cited text no. 28
    29.Du H, Hirabayashi K, Ahn S, Kren NP, Montgomery SA, Wang X, et al. Antitumor responses in the absence of toxicity in solid tumors by targeting B7-H3 via chimeric antigen receptor T cells. Cancer Cell 2019;35:221-37.  Back to cited text no. 29
    30.Lin CW, Wang YJ, Lai TY, Hsu TL, Han SY, Wu HC, et al. Homogeneous antibody and CAR-T cells with improved effector functions targeting SSEA-4 glycan on pancreatic cancer. Proc Natl Acad 2021;50:e2114774118.  Back to cited text no. 30
    31.Teng KY, Mansour AG, Zhu Z, Li Z, Tian L, Ma S, et al. Off-the-shelf prostate stem cell antigen-directed chimeric antigen receptor natural killer cell therapy to treat pancreatic cancer. Gastroenterology 2022;22:1-4.  Back to cited text no. 31
    32.Jan CI, Huang SW, Canoll P, Bruce JN, Lin YC, Pan CM et al. Targeting human leukocyte antigen G with chimeric antigen receptors of natural killer cells convert immunosuppression to ablate solid tumors. Immunother Cancer 2021;10:e003050.  Back to cited text no. 32
    33.Xia N, Haopeng P, Gong JU, Lu J, Chen Z, Zheng Y, et al. Robo1-specific CAR-NK immunotherapy enhances efficacy of 125I seed brachytherapy in an orthotopic mouse model of human pancreatic carcinoma. Anticancer Res 2019;1:5919-25.  Back to cited text no. 33
    34.Lee YE, Ju A, Choi HW, Kim JC, Kim EE, Kim TS, et al. Rationally designed redirection of natural killer cells anchoring a cytotoxic ligand for pancreatic cancer treatment. J Control Release 2020;326:310-23.  Back to cited text no. 34
    35.Zeng W, Zhang P. Resistance and recurrence of malignancies after CAR-T cell therapy. Exp Cell Res 2022;2:112971.  Back to cited text no. 35
    36.Hu J, Sun C, Bernatchez C, Xia X, Hwu P, Dotti G, et al. T-cell homing therapy for reducing regulatory T cells and preserving effector T-cell function in large solid tumors. Clin Cancer Res 2018;24:2920-34.  Back to cited text no. 36
    37.Jin L, Tao H, Karachi A, Long Y, Hou AY, Na M, et al. CXCR1- or CXCR2-modified CAR T cells co-opt IL-8 for maximal antitumor efficacy in solid tumors. Nat Commun 2019;1:4016. doi: 10.1038/s41467-019-11869-4.  Back to cited text no. 37
    38.D'Aloia MM, Zizzari IG, Sacchetti B, Pierelli L, Alimandi M. CAR-T cells: The long and winding road to solid tumors. Cell Death Dis 2018;9:282.  Back to cited text no. 38
    39.Nakamura T, Kawakami K, Nomura M, Sato Y, Hyodo M, Hatakeyama H, et al. Combined nano cancer immunotherapy based on immune status in a tumor microenvironment. J Control Release 2022;17:S0168-3659.  Back to cited text no. 39
    40.Hao Y, Li H, Zhao H, Liu Y, Ge X, Li X, et al. An intelligent nano-vehicle armed with multi-functional navigation for precise delivery of toll-like receptor 7/8 agonist and immunogenic cell death amplifiers to eliminate solid tumors and trigger durable antitumor immunity. Adv Healthc Mater 2022;19:e2102739.  Back to cited text no. 40
    41.Xia H, Qin M, Wang Z, Wang Y, Chen B, Wan F, et al. A pH-/Enzyme-Responsive Nanoparticle Selectively Targets Endosomal Toll-like Receptors to Potentiate Robust Cancer Vaccination.Nano Lett 2022 ;7:2978-2987.  Back to cited text no. 41
    42.Miller IC, Zamat A, Sun LK, Phuengkham H, Harris AM, Gamboa L, et al. Enhanced intratumoural activity of CAR T cells engineered to produce immunomodulators under photothermal control. Nat Biomed Eng 2021;11:1348-59.  Back to cited text no. 42
    43.Kavanag HH, Dunne S, Mcfadde NE. Human CAR-T cells engineered using the solupore ex vivo cell engineering platform are highly cytotoxic and specific against CD19+cells in vitro. Cytotherapy 2020;5:S29.  Back to cited text no. 43
    44.Luo H, Wu X, Sun R, Su J, Wang Y, Dong Y, et al. Target-dependent expression of IL12 by synNotch receptor-engineered NK92 cells increases the antitumor activities of CAR-T cells. Front Oncol 2019;9:1448.  Back to cited text no. 44
    45.Ali AI, Oliver AJ, Samiei T, Chan JD, Kershaw MH, Slaney CY. Genetic redirection of T cells for the treatment of pancreatic cancer. Front Oncol 2019;9:56.  Back to cited text no. 45
    46.Mansouri V, Yazdanpanah N, Rezaei N. The immunologic aspects of cytokine release syndrome and graft versus host disease following CAR- T cell therapy. Int Rev Immunol 2021;5:1-20.  Back to cited text no. 46
    47.Totzeck M, Michel L, Lin Y, Herrmann J, Rassaf T. Cardiotoxicity from chimeric antigen receptor-T cell therapy for advanced malignancies. Eur Heart 2022;8:ehac106.  Back to cited text no. 47
    48.Shi YG, Yu YH. Current status and outlook of chimeric antigen receptor T cell therapy causing cytokine release syndrome and neurotoxicity. Practical Medicine 2021;2:268-71.  Back to cited text no. 48
    49.Liu Y, Liu G, Wang J, Zheng ZY, Jia L, Rui W, et al. Chimeric STAR receptors using TCR machinery mediate robust responses against solid tumors. Transl Med 2021;586:eabb5191.  Back to cited text no. 49
    50.Weber EW, Parker KR, Sotillo E, Lynn RC, Anbunathan H, Lattin J, et al. Transient rest restores functionality in exhausted CAR-T cells through epigenetic remodeling. Science 2021;6537:eaba1786.  Back to cited text no. 50
    51.Ye L, Park JJ, Peng L, Yang Q, Chow RD, Dong MB, et al. A genome-scale gain-of-function CRISPR screen in CD8 T cells identifies proline metabolism as a means to enhance CAR-T therapy. Cell Metab 2022;34:1-20.  Back to cited text no. 51
    52.Dai H, Wu Z, Jia H, Tong C, Guo Y, Ti D, et al. Bispecific CAR-T cells targeting both CD19 andCD22 for therapy of adults with relapsed or refractory B cell acute lymphoblastic leukemia. J Hematol Oncol 2020;1:30.  Back to cited text no. 52
    53.Park HR, Jung U. Depletion of NK Cells Resistant to Ionizing Radiation Increases Mutations in Mice After Whole- body Irradiation. In Vivo 2021;3:1507-1513.  Back to cited text no. 53
    54.Zhang S, Yu F, Che A, Tan B, Huang C, Chen Y, et al. Neuroendocrine Regulation of Stress-Induced T Cell dysfunction during Lung Cancer Immunosurveillance via the Kisspeptin/ GPR54 Signaling Pathway. Adv Sci 2022; 27:e2104132.  Back to cited text no. 54
    55.Alishah K, Birtel M, Masoumi E, Jafarzadeh L, Mirzaee HR, Hadjati J, et al. CRISPR/Cas9-mediated TGFβRII disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells in vitro. J Transl Med 2021;1:482.  Back to cited text no. 55
    56.Daher M, Basar R, Gokdemir E, Baran N, Uprety N, Nunez Cortes AK, et al. Targeting a cytokine checkpoint enhances the fitness of armored cord blood CAR-NK cells. Blood 2020;5:624-36.  Back to cited text no. 56
    57.Gurney M, Stikvoort A, Nolan E

留言 (0)

沒有登入
gif