Skip to main content

Infusion and delivery strategies to maximize the efficacy of CAR-T cell immunotherapy for cancers

Abstract

Chimeric antigen receptor (CAR) T-cell therapy has achieved substantial clinical outcomes for tumors, especially for hematological malignancies. However, extending the duration of remission, reduction of relapse for hematological malignancies and improvement of the anti-tumor efficacy for solid tumors are challenges for CAR-T cells immunotherapy. Besides the endeavors to enhance the functionality of CAR-T cell per se, optimization of the infusion and delivery strategies facilitates the breakthrough of the hurdles that limited the efficacy of this cancer immunotherapy. Here, we summarized the infusion and delivery strategies of CAR-T cell therapies under pre-clinical study, clinical trials and on-market status, through which the improvements of safety and efficacy for hematological and solid tumors were analyzed. Of note, novel infusion and delivery strategies, including local-regional infusion, biomaterials bearing the CAR-T cells and multiple infusion technique, overcome many limitations of CAR-T cell therapy. This review provides hints to determine infusion and delivery strategies of CAR-T cell cancer immunotherapy to maximize clinical benefits.

Introduction

Genetically engineering T cells to express CAR molecules has significantly boosted the advancement of celluar immunotherapy [1]. As a living drug, CAR-T cells are manufactured ex vivo for sufficient expansion and then reinfused into the patient to exert tumor-targted killing activity against tumor cells. CAR-T cell therapy has shifted the paradigm for the treatment of cancer and has become one of the mainstream treatments for refractory and relapsed lymphoma [2]. Currently, six CAR-T cell products for hematologic malignancies have been approved by the American Food and Drug Administration (FDA), achieving impressive clinical outcome in multi-line therapy-refractory patients [3].

Systemic injection of CAR-T cells to patients with hematological tumors has demonstrated feasible and effective [4]. Post-infusion CAR-T cells can circulate throughout the body and dynamically search antigen-specific tumor cells. Upon recognizing target antigen, CAR-T cells are activated to proliferate and exert robust tumor lysis ability [5]. However, poor persistence of CAR-T cells closely correlated with the unsatisfactory outcomes and disease replase in patients receiving CAR-T therapy [6]. Investigators have been endeavoring to develpe various gene-engineering strategies to ameliorate exhaustion and augment the persistence of CAR-T cells, thus improving the duration time of anti-tumor efficacy. From the perspective of clinical administration scheme of CAR-T therapy, the cell-infusion strategy is also one of decisive factors of CAR-T therapeutic effectiveness.

Cytokine release syndrome (CRS) and neurotoxicity are the major side effects in CAR-T cell immunotherapy [7]. High infusion dose of CAR-T cells is identified as one of the main factors causing severe CRS [8, 9]. According to the data disclosed in Clinical trials.gov, a varied range of CAR-T cell doses have been investigated to balance the safety and efficacy of CAR-T cell therapy. In addition to considering the total infusion dose, fractional infusion of a total dose provides a strategy to flexibly adjust total CAR-T infusion dose and make the risk of adverse effects more controllable [1, 10].

Based on the success of CAR T-cell therapy in hematological malignancies, researchers have ventured into expanding this therapeutic modality to address solid tumors [11]. However, the concealed location of solid tumors and immunosuppressive microenvironment pose significant barriers, limiting the therapeutic efficacy in treating patients with solid tumors [12, 13]. In terms of CAR-T delivery strategy, the approach differs significantly from that employed in hematological malignancies. Through systemic infusion that is used in treating hematological tumors, CAR-T cells need to circulate, migrate, and break the anatomical barriers to reach the tumor site [14]. During this journey, a portion of CAR-T cells are distributed to different tissues and experience activation-induced mortality within circulation. These conditions can contribute to deficient quantity and quality of CAR-T cells, significantly hampering their anti-tumor activities. Therefore, to overcome these challenges, locoregional delivery strategies such as intraperitoneal injection [15, 16], intrathoracic injection [17], hepatic artery injection, catheter injection [18] have been explored to treat a series of solid tumors. While intravenous (i.v) injection of CAR-T cells remains a prevalent method to treat solid tumors [19, 20], which typically requires multiple doses to achieve sufficient effectiveness [21, 22]. Furthermore, innovative adjunctive delivery methods utilizing biomaterials have been developed to boost CAR-T performance [23]. These advanced strategies harness the unique properties of biomaterials to improve CAR-T cell survival, trafficking, and tumor infiltration, thereby potentiating their anti-tumor efficacy. By combining with these interdisciplinary innovations, it is promising to overcome the challenges existing in traditional CAR-T cell delivery mode and unleash the maximal efficacy of CAR-T therapy.

In this review, we mainly focus on the researches regarding infusion dose and delivery strategies of CAR-T cells in treating hematological and solid tumors. Through summarizing the datas and findings disclosed in preclinical and clinical studies, we give insights into choosing proper infusion dose and delivery strategy of CAR-T cells while taking safety and efficacy into account. In addition, we emphasize on novel infusion and delivery techniques including locoregional infusion method, biomaterials-based delivery system and multiple infusion modality, holding great potential to overcome part of the limitations in traditional CAR-T cell therapy. Overall, through a better understanding of the latest CAR-T infusion and delivery strategies, we hope to offer guidance on how to optimize CAR-T cell infusion dose and delivery modality to maximize their clinical benefits for patients with cancer.

Infusion strategies of CAR-T cell therapy for hematological malignancies

Prior to CAR-T cell infusion, lymphodepletion is a necessity to enable effective and durable therapeutic responses [24]. The common lymphodepletion regimens include cyclophosphamide, fludarabine, bendamustine, azacytidine [25, 26]. In Authority-approved CAR-T cell products CAR-T cell therapies, lymphodepletion chemotherapy typically employs a combination of cyclophosphamide and fludarabine [27]. This regimen can effectively decrease the circulating immune cells, thereby facilitating optimal proliferation and anti-tumor activity of CAR-T cells [28]. We summarized the components, usual doses and schedules of lymphodepletion regimens. At the same time, some common lymphodepletion drugs’ information in clinical trials is also listed (Table 1). After a few days of lymphodepletion, CAR-T cells are intravenously injected into the body and travel in the vascular system. Part of the CAR-T cells can rapidly egress from circulatory system and reside in different tissues, resulting in a quick quantity decrease of circulating CAR-T cells. Upon recognizing the tumor mass, CAR-T cells can be activated to proliferate and alter biodistribution to search for cognate antigens [29]. This long journey can lead to exhaustion, poor persistence and ultimately, unsatisfactory therapeutic efficacy of CAR-T cells. Clinically, to address these challenges, lymphokines/cytokines are often administered to prolong the lifespan and activity of CAR-T cells. Additionally, a wide range of CAR-T cell doses have been explored, spanning one or more orders of magnitude. Next, we focus on summarizing the clinical infusion schemes of CAR-T cells for hematological tumors [30,31,32,33,34].

Table 1 Lymphodepletion strategies commonly used in current clinical trials

The infusion dose of CAR-T cells for clinical use

The success of CAR-T cell therapy is evidenced by authority-approved CAR-T cell products for different hematological malignancies. The infusion dose of the CAR-T products for different indications varies (Table 2). According to www.clinicaltrials, we calculated and summarized the disclosed infusion dose or dose range of CAR-T therapies.

Dose selection is a key point of the success of CAR-T cell therapy. The infusion dose of CAR-T cells is strongly associated with CRS and immune effector cell-associated neurotoxicity syndrome (ICANS) [37]. In the majority of the trials, the infusion doses of CAR-T cells are below 3 × 106 cells/kg for hematological malignancies (Fig. 1). This general threshold of infusion dose may represent as an approximate safe dose, above which may induce adverse effect and uncontrollable outcome [38]. Some studies reported that higher dose levels of CAR-T cells may cause severe toxic effects [39,40,41]. In a case report, a patient with multiple myeloma was observed the neurotoxicity post high infusion dose of CAR-T cells [42]. In another clinical trial of CD19 CAR-T cells in patients with acute lymphoblastic leukemia, 3 CRS-related deaths observed after CD19 CAR-T cells were infused with a high dose [43]. To mitigate the adverse effect of CAR-T therapy, numerous approaches have been explored in clinical practice. The administration of anti-IL-6 receptor antibody tocilizumab has proven effective in managing CRS, with corticosteroids serving as an additional line of treatment for severe CRS cases [44]. ICANS generally occurs after the symptoms of CRS have subsided. In case of neurologic toxicity, hormonal management is the initial choice due to the inability of monoclonal antibodies rapidly cross the blood-brain barrier. Low-grade ICANS is typically managed by supportive care, whereas severe ICANS is usually treated with corticosteroids [45]. “On-target, off-tumor” also poses a potentially fatal risk in CAR-T therapy. Recently, “suicide genes” including inducible caspase 9 and truncated version of EGFR were incorporated to CAR-T cells to overcome the obstacle [46]. A clinical study published in 2023 demonstrated that the safety performance was enhanced through the use of inducible caspase 9 suicide in patients receiving GD2-CAR-T cells (NCT03373097) [47]. However, irreversible elimination of CAR-T cells by suicide gene prior to eradicate tumor completely might limit clinical efficacy [48]. A potential strategy is designing reversible off/on-switches, which permits CAR-T cell switch between “on” and “off” states. Progress has been made in the administration of some small molecules such as fluorescein isothiocyanate, folate, rimiducid, rapamycin, and proteolysis-targeting chimera compounds [49].

Table 2 Authority-approved CAR-T cell products

The infusion amount of CAR-T cells in different researches varies significantly due to several factors, including the choice of tumor targets, costimulatory domains, and manufacture processes. It has been identified that 4-1BB-incorporated CAR-T cells have superior persistence and less neurological toxicity, compared with CD28 counterparts in clinical tests [50, 51]. Therefore, the choice of CAR-T infusion dose is crucial in determining the efficacy and safety profile of the treatment when it comes to different CAR-T products.

Fig. 1
figure 1

Number of CAR-T clinical trials in different infusion dose intervals. We divide the infusion doses into four dose-intervals. The median value of CAR-T infusion dose range in each clinical trial is calculated and the number of clinical trials in each dose interval is counted. The number of clinical trials is respectively counted in hematologic and solid tumors. The data is summarized according to clinical trials.gov

CD19-targeted CAR-T cell therapies are the most widely studied in clinical trials. Four CD19-targeted CAR-T products have been approved by FDA for B cell-derived lymphoma and leukemia. The infusion doses of these products are wide-ranging. Taking the tisa-cel for example, adolescents up to 25 years of age receive the low dose (10–250 million CAR-T cells), and the higher dose (60–600 million cells) is suitable for adults (Table 2). However, the lack of transparency in clinical trial reporting, particularly regarding patient information and specific CAR-T cell varieties used, can make it challenging to gain a comprehensive understanding of the dose landscape for CAR-T cell therapies. Nonetheless, by summarizing the dose ranges reported in a mass of studies, we can gain some insights into the general map of CAR-T cell infusion pattern. There are a total of 122 clinical registry trials that have exposed doses of CD19 CAR-T cell therapies. The dose range for the treatment of hematological malignancies is mostly from 1 × 106 to 1 × 107cells/kg (Fig. 2A). A positive correlation between therapeutic response and infusion dose levels was reported in some studies [39, 52, 53]. In a phase I trial of CAR-T cell therapy for B-cell lymphoma, patients received a single intravenous infusion at a high dose of 2 × 106 CAR-T cells/kg, the objective response rate(ORR) was 82%, and the complete response rate was 54% [54]. In another phase I trial, bispecific anti-CD20/CD19 CAR T cells for the treatment of recurrent B-cell malignancies were administrated in dose-escalating way, ranging from 2.5 × 105 to 2.5 × 106 cells/kg. The results show the ORR was 100% at the infusion dose of 2.5 × 106 cells/kg (CR was 92% and partial response (PR) was 8%) [55]. It has been reported that CAR-T cell infusion dose has a threshold, beneath which the infusion dose has a positive correlation with the clinical outcome of CAR-T cells. When surpassing this threshold, the clinical response of CAR-T therapy may peak and reach a plateau [38, 56]. Notably, studies involving anti-CD19 CAR-T cells have demonstrated optimal clinical efficacy at doses typically lower than 150 million cells [57,58,59,60].

BCMA-targeted CAR-T cell therapies have been approved in the United States for treating multiple myeloma due to their high safety and efficacy. In clinical trials, the dose range of BCMA-targeted CAR-T cells for the treatment of relapsed/refractory multiple myeloma mostly covered the range from 0.5 × 106 cells/kg (NCT03672253) to 5 × 106 cells/kg (NCT04194931) (Fig. 2A). BCMA-targeted CAR-T cell studies may need higher doses [61,62,63] to achieve optimal clinical efficacy than CD19-targeted CAR-T cells. In a clinical trial, 16 patients received 9 × 106 BCMA-targeted CAR-T cells/kg at the highest dose, attaining 81% ORR and 63% good PR or CR [64].

Transmembrane glycoprotein CD7 is an attractive target in T cell malignancies since it is expressed in over 95% of leukemia and lymphoma produced from T cells [65]. To date, there are 41 clinical trials of CAR-T cell therapies targeting CD7, and most of them cover doses ranging from 0.5 × 106 (NCT04840875) to 6 × 106cells/kg (NCT05127135) (Fig. 2A). A phase I clinical trial was conducted to test genetically modified CD7-targeted allogeneic CAR-T cell therapy in hematologic malignancies. The trial used a dose-escalation design with three levels (level 1: 1 × 107cells/kg; level 2: 2 × 107cells/kg; level 3: 3 × 107cells/kg) to evaluate the safety and tolerability of CD7-targeting CAR-T cells, 81.8% of patients showed objective responses and the CR rate was 63.6% [66]. However, since CD7 is expressed on most T cells, CD7 antigen-specific CAR-T cells can produce severe suicide during preparation. Various techniques including gene editing, protein blockers, and natural selection have been explored to overcome challenges and enhance the capabilities of CD7 CAR-T to lysis T-lymphocyte [67].

CD30, is a type of cell surface glycoprotein that is highly expressed on the surface of Hodgkin’s lymphoma, anaplastic large cell lymphoma, and other lymphoma cells. It is important to note that the expression of CD30 is very low or non-existent on the surface of normal cells and tissues [68]. CAR-T cells targeting CD30 have shown high response rates and low toxicity in patients with relapsed/refractory CD30+ hematologic malignancies, particularly in classical Hodgkin lymphoma [69]. To date, an increasing number of CD30 targeting CAR-T cells have been registered in clinical trials, most of which define infusion unit of CAR-T cells by body surface area or total cells. To compare doses across studies, we normalized doses by calculating 70 kg of body weight or 1.6 m2 of body-surface area. It was found that CD30 CAR-T dose range mostly covered between 106 to 108cells/kg (Fig. 2A). A research from Baylor College of Medicine and the University of North Carolina showed that autologous CD30 CAR-T cell therapy had a high CR, durability, and a favorable safety profile. Two phase 1/2 trials (NCT02690545 and NCT02917083) involved 41 patients with relapsed/refractory Hodgkin’s lymphoma receiving CD30-targeting CAR-T cells. An expansion cohort of patients at both institutions received the highest dose level of 2 × 108 CAR-T cells/m2. The results showed that CD30-targeting CAR-T therapy showed superior efficacy than conventional CAR-T therapy in the treatment of patients with relapsed/refractory Hodgkin lymphoma [70].

The field of CAR-T cell therapy for hematological malignancies has been rapidly expanding, with researchers exploring various novel targets beyond the classical ones like CD19, BCMA, CD7, and CD30 (Fig. 2A). We also provide a valuable overview of the dose ranges being explored for some of these emerging targets in CAR-T cell therapy and listed in Table 3.

Table 3 Overview of CAR-T infusion dose of hematological malignancies in clinical studies (not depicted in Fig. 2)
Fig. 2
figure 2

The dose distribution of CAR-T cells on different targets for hematological malignancies and solid tumors in clinical trials. The data is summarized according to clinical trials.gov. Each bar represents the CAR-T infusion dose/dose range in a clinical trial(A. Summarized clinical trials data for hematological malignancies. B. Summarized clinical trials data for solid malignancies) to uniformly compare the infusion dose of CAR-T cells across clinical studies, we normalize the CAR-T dose unit at 106 cells/kg (calculated for 70 kg/patient or 1.6 m2/patient if the dose was not flat). The data is ranked in increasing order of CAR-T max dose in each target. ( represents three or more than three clinical trials adopting the same CAR-T dose/dose range)

Dose-split strategy of CAR-T cells infusion

The activation of CAR-T cells is a complex process that can lead to the release of inflammatory cytokines, including interferon and tumor necrosis factor. These substances can trigger the release of additional cytokines from macrophages and monocytes, leading to endothelial damage, CRS and ICANS events [71]. The infusion dose of CAR-T cells, the kinetics of CAR-T cell expansion and tumor burden are the major factors affecting the severity of CRS [8, 9]. Patients with a high tumor burden have been identified with a higher risk of CRS [72, 73]. It has been suggested that CAR-T cell dose fractionation or split dosing can reduce the release of inflammatory cytokines and address the CRS issue [1]. Indeed, in studies using split dosing of CAR-T cells, the incidence of grade 3 or higher CRS can be significantly reduced. Frey NV split the total dose of 500 million CAR-T cells into 10%, 30%, and 60% and infused in the first three days for the treatment of acute lymphocytic leukemia. The results of the study showed that the incidence of CRS in the dose-graded group was significantly lower than that in the low single-dose group [43]. This split dosing strategy has also achieved good clinical results in CD19 CAR-T cells for chronic lymphocytic [26, 74, 75]. In addition to the above dose-split protocol, Xu J et al. divided the total dose into 33%, which was injected on day 0, on day 3, and on day 6 [76]. Some scientists even split the total dose into two (33% and 67%) and infused on the first two days. The results showed that this delivery strategy significantly reduced CRS and improved the safety of CAR-T cell therapy [77, 78]. In patients with high tumor burden, lowering the infusion dose reduces peak cytokine levels and the severity of CRS. However, lowering the dose may also result in an insufficient tumor-lysis, resulting in incomplete clearance of all tumor cells. Thus, the administration of CAR-T cells with a dose-splitting strategy can stagger the rise in cytokine levels, resulting in a lower peak that decreases the severity of CRS.

Infusion of fresh CAR-T cells vs. cryopreserved CAR-T cells

CAR-T cells are usually cryopreserved to facilitate the completion of rigorous quality control tests and enable flexible infusion schedule based on the patient’s physical condition [79]. However, some studies have found that freshly made CAR-T cells have more potential and advantages compared to cryopreserved ones [80]. Shah et al. observed that patients who received fresh CD20/CD19 tandem bispecific CAR-T cells had increased peak CAR-T cell expansion levels and ORR compared to patients infused with cryopreserved CAR-T cells [55]. Nonetheless, cryopreservation has minimal effect on the fundamental characteristics of CAR-T cells. Studies have consistently demonstrated that the survival rate of resuscitated CAR-T cells following cryopreservation remains high, exceeding 80% [81,82,83,84]. In a study published in 2018, the transduction rates of cryo-thawed CAR-T cells from three healthy donors were examined and found no statistically significant differences compared to their pre-cryopreservation counterparts(41.9% vs. 43.5%; 68.3% vs. 69%; 37% vs. 37.3%, P > 0.05) [82]. Similar conclusions have been drawn in other studies [83, 84]. Furthermore, cryopreservation has been identified to have negligible effects on the final CAR-T cell composition, as evidenced by the account of CD3 positive cell population(98% ± 2.1% vs. 98% ± 2.4%)and the ration of CD4 and CD8 T cells(2.2 ± 3.9 vs. 2.3 ± 4.0) following resuscitation [83, 85, 86]. Results from a clinical trial comparing the infusion of fresh and cryopreserved targeted CD19 CAR-T cells in Non-Hodgkin’s lymphoma patients showed that the cryopreserved group had a lower rate of acute hematological toxic events compared to the fresh group. One possible reason for the different safety profiles lies in that the quality control parameters of cryopreserved CAR-T cells could remain consistent during transportation from good manufacturing practice facility to the hospitals, but the variant of fresh CAR-T cell parameters was relatively higher [79].

Currently, cryopreserved CAR-T products are still widely employed in clinical trials due to their many advantages over fresh CAR-T cells. Central manufacturing facilities can more easily control the cryopreserved CAR-T cell quality. Additionally, cryopreserved formulations are more cost-effective since they do not require the repeated manufacturing of fresh products for each patient, allowing for more efficient infusion scheduling and patient management [87, 88]. A small number of studies have investigated the effects of cryopreservation on CAR-T cells, more investigations are needed to fully understand the potential impact of this process on cell function and efficacy. Furthermore, based on the convenience and potential of cryopreserved cell product, we should put an emphasis on optimizing the cryopreservation process and to determining the optimal conditions for storing and transporting these cells to ensure the best possible outcomes for patients.

CAR-T cells delivery strategies of solid tumors

Systematic infusion of CAR-T cells to patients with hematological tumors has achieved encouraging efficacy. Nevertheless, intravenous infusion of CAR-T cells to patients with solid tumors has not replicated the identical success due to the different physical and physiological attributes [89]. Considering these challenges, increasing the intravenous infusion dose and optimizing CAR-T infusion scheme are of significant necessity to ensure the effectiveness of CAR-T cells in solid tumors [90]. Take the hepatic tumor for example, it is desirable to conduct hepatic artery injection to control the volumetric blood flow rate at a low level [91]. Solid tumors grow in concealed locations of the body and form complex tumor microenvironment (TME) such as extracellular matrix (ECM), tumor vasculature, fibroblasts, and immune-suppressive substances, hindering the trafficking and migration of CAR-T cells to solid tumor beds [92]. Therefore, more investigators adopt locoregional infusion methods to deliver CAR-T cells into tumor tissue, which presents as a feasible therapeutic strategy to improve the trafficking, infiltration and efficiency of CAR-T cells. Direct regional injection can avoid the consumption and exhaustion of CAR-T cells during long circulating journey to tumor sites. Due to a more concentrated distribution of CAR-T cells around the tumor bed, off-target and/or dose-related toxicities could be mitigated as well [93, 94].

The infusion dose of CAR-T cells for clinical use in solid malignancies

Compared to hematological malignancies, the clinical investigation and progression of solid tumors are relatively limited. Investigators have been attempting to target more antigens such as MSLN, HER2, EGFR, GPC3, and Claudin 18.2 to expand the curative potential of CAR-T cells. The tumor antigens targeted by CAR-T products in clinical trials are numerous but few studies have published detailed curative schemes. Since there is little consensus on the number and frequency of CAR-T cells infusion [95], we summarized the infusion dose range of CAR-T cells applied in clinical trials according to the classification of targets. In summary, the infusion dose range of CAR-T cells in clinical trials for solid tumors varies widely, with most doses ranging from 105 to 108 cells per kilogram of body weight (Fig. 2B).

Regional delivery strategies in different parts of body

Treating solid tumors by CAR-T cell therapy has garnered significant scientific and clinical attention in recent years. Solid tumor clumps tend to be surrounded with abundant tumor-associated fibroblasts and blood vessels [96], forming physical barriers to prevent CAR-T cells from penetrating into the interior of tumor site [97]. In addition, immunosuppressive TME directly impact on the clinical efficacy of CAR-T cell therapy [98]. These factors pose significant challenges for the market translation of CAR-T therapy treating solid tumors. Regional delivery of CAR-T cells has been demonstrated to be safe and feasible in solid tumors [99, 100]. The delivery strategy can promote invasion, proliferation, trafficking, and stimulate functionally sustained systemic immunity. CAR-T cells can be delivered regionally to tumor sites with sustained function. Including intra-tumoral injection, arterial infusion, intraperitoneal injection, and intraventricular injection (Fig. 3).

Fig. 3
figure 3

Delivery strategies of CAR-T cells in clinical setting. Intravenous infusion is the major delivery method in treating patients with hematological tumors. Due to the anatomical barrier and TME of solid tumors, multiple locoregional delivery methods have developed for specific tumors. We counted the percent of clinical trials adopting different delivery strategies

Intratumoral injection

Intratumoral injection can increase CAR-T cell bioavailability inside tumors, enhance the efficacy of immunotherapies and reduce systemic toxicities [101]. Intratumoral injection does not cause direct normal tissue damage compared with resection or radiation. This delivery method is more suitable for visible or palpable tumors, such as melanoma [102, 103]. By the guidance of ultrasound and computed tomography (CT), CAR-T cells can be intratumorally infused in unresectable or medically inoperable tumors [104]. Wang et al. discovered that intratumoral injection of CAR-T cells could eradicate tumors, whereas intravenous injection could only inhibit tumor growth [105]. The injection dose depends on the interstitial pressure and size of the tumor. For refractory and relapsed tumor, multiple intratumoral injections might be needed to stimulate the antitumor immune response [93]. Although repeated punctures on tumor clump can lead to organ damage and the risk of tumor metastasis, clinical outcomes have confirmed the strength of intertumoral injection outweigh its defects in patients whose disease condition have been assessed [106, 107].

To date, there have been 14 clinical trials that have attempted direct intratumoral injection of CAR-T cells (Fig. 3). In a clinical trial of CAR-T cells to treat metastatic breast cancer, patients received a single intratumoral injection of 3 × 107 or 3 × 108 cells. The results showed intratumoral injection of CAR-T cells was well tolerated in all 6 patients. CAR-T mRNA was detectable in peripheral blood and the injected tumor tissue [106]. In a clinical trial published in 2023 for squamous-cell carcinoma of the head and neck, fifteen subjects were treated across five dose cohorts ranging from 1 × 107 to 1 × 109 autologous cells under the guidance of ultrasonography, 60% of subjects obtained disease control, with no treatment-related adverse events above grade 2 were observed [108]. These trials confirmed that intratumoral administration was safe and feasible. This approach could largely reduce systemic toxicities and adverse events since the main immune responses occur locally. Additionally, this approach can combine with other systemic therapies without adding more toxicities [101, 109].

Arterial infusion

Intra-arterial delivery is another potential delivery strategy for the regional administration of CAR-T cells [110]. Combining pressure-enabled drug delivery technology with hepatic arterial infusion of CAR-T cells can overcome excessive intra-tumoral pressure and enhance delivery efficiency [111]. Before CAR-T cell infusion, a mapping angiogram was performed via a common femoral artery approach. Next, extrahepatic sites such as the gastroduodenal and right gastric arteries were embolized with microcoils to conduct CAR-T perfusion. Post CAR-T cells injection at a specific speed via a syringe, angiography with a calibrated contrast rate was performed to confirm preserved arterial flow [112]. To date, there have been a limited number of clinical trials that have attempted direct arterial injection of CAR-T cells (Fig. 3). This delivery strategy has been used more frequently in digestive system malignancies. Katz et al. reported their phase I study of local intrahepatic CAR-T cells in the treatment of malignant tumors with liver metastasis, three patients received anti-CEA CAR-T cells through hepatic arterial infusion in dose escalation manner. The results have proved the safety of arterial infusion CAR-T cells [112]. Hepatic arterial infusion of CAR-T cells has also been used in the treatment of colorectal cancer, even receiving a high dose of 1 × 1010 CEA CAR-T cells through hepatic arterial infusion, patients with pancreatic cancer did not undergo serious adverse events above grade 3 or on-target/off-target. Compared with the median survival time of 5 months in patients who experienced intravenous injection, the overall survival time of a patient receiving hepatic arterial infusion significantly pronged, up to 23.2 months [113, 114].

Intraperitoneal and intrapleural injection

In the past 20 years, intraperitoneal and intrapleural injection of drugs have been mainly used for cancer chemotherapy and achieved good clinical results [115,116,117]. In recent years, increasing interests have been focused in adopting intraperitoneal and intrapleural delivery strategy to infuse CAR-T cells to solid tumors, showing inspiring efficacy and safety [118,119,120]. Intraperitoneal infusion have beneficial effect in tumor cells that have unique patterns of spread over the serosal surface [121]. Regional intrapleural and intraperitoneal administration can help increase efficacy and persistence by delivering cells directly into the tumor [122]. In a study that treating epithelial ovarian cancer with ErbB2-targeting CAR-T cells, researchers found that intraperitoneal infusion CAR-T cells offered a safer and more effective strategy than intravenous treatments. The results of this study demonstrated that tumor-bearing mice treated with CAR-T cells by intraperitoneal infusin achieved disease remission and increased survival period compared with intravenous infusion [123]. Additionally, intraperitoneal and intrapleural delivery strategy have also demonstrated potential in clinical stage for treating solid tumors. There have been 16 registered CAR-T clinical trials using intraperitoneal and intrapleural injection to treat solid tumors (Fig. 3), such as malignant pleural mesothelioma (NCT04577326), ovarian cancer (NCT05211557), and pancreatic cancer (NCT03323944). In one phase I trial, a single dose of 1 × 106 CAR-T cells targeting fibroblast-activating protein were delivered to pleural of patients with pleural mesothelioma [124]. The results demonstrated that CAR-T cells indicated an ongoing immune response with a high safety profile in vivo. Intraperitoneal delivery was also utilized in a phase I dose-escalation trial against ovarian cancer and peritoneal mesothelioma. CAR-T cells were injected weekly for 3 weeks (NCT03608618). This study’s preliminary results showed that the treatments were well tolerated, 4 out of 11 patients showed initial stable disease, and 3 patients were in a stable condition for more than 2 months. Combining intravenous with intrapleural injection to deliver CAR-T cells is also a strategy for the treatment of abdominal malignant tumors. In a standard 3 + 3 dose-escalation phase I trial, patients were infused with escalating doses of CAR-T cells from 3 × 105 to 1 × 107 cells/kg to establish the maximum tolerated dose (MTD). All patients will receive 50% of the genetically CAR-T cell dose intravenously, the remaining dose of cells will be administered by intrapleural infusion 3 days later [125].

Intraventricular injection

Intraventricular administration of CAR-T cells to target central nervous system (CNS) tumors has shown promising preclinical and early clinical results [22, 90, 126, 127]. In a preclinical study, CAR-T cells were injected intracranially to treat the malignant glioma. Kiwan Kim et al. found that the tumor volume was significantly reduced in tumor-bearing mice and the survival rate of the mice was markedly improved [128]. Infusing CAR-T cells with the assist of intracranial catheter has been demonstrated the efficiency and safety [94, 129]. Nicholas A. Vitanza et al. first reported the efficacy of repeated intracranial B7-H3 CAR-T cells for patients with diffuse intrinsic pontine glioma. The data from this trial suggested the feasibility of repeated intraventricular injection of B7-H3 CAR T-cells, which can induce local immune activation [130]. Clinical trials for the treatment of CNS tumors with different targets can also achieve similar efficacy through intraventricular injection of CAR-T cells. HER2-specific CAR-T cells were repeatedly administered by intraventricular injection to children and young adults with recurrent or refractory HER2-expressing CNS tumors at doses ranging from 1 × 107 to 1 × 108 cells [131]. In March 2023, investigators described the successful intraventricular administration of 1 × 105cells/kg of GD2-Specific 4SCAR-T cells in patients with glioblastoma. 4 of the 8 evaluable patients showed a PR for 3 to 24 months, 1 patient had a stable disease condition for 4 months after infusion [132]. CAR-T cells administered intraventricularly to treat cerebral tumors exhibited faster kinetics, greater potency, and reduced systemic levels of inflammatory cytokines compared with CAR-T cells administered intravenously [90].

In the past 5 years, the number of registered clinical trials exploring locoregional delivery of CAR-T cells in solid tumors has grown considerably. CAR-T cell therapy offers a way to circumvent normal-tissue, on-target, off-tumor toxicity [48]. It allows more concentrated density of CAR-T cells in the solid tumor bed to enhance anti-tumor activity. Significant and durable clinical response have further stimulated the investigators’enthusiam in the advancement of novel regional delivery strategy [99].

Novel adjunctive delivery strategies of CAR-T cells at the preclinical stage

Effective anti-tumor responses require CAR-T cells to be highly activated and persistent at the tumor site [133]. Though locoregional delivery can augment the penetration and viability of CAR-T cells, the lack of sustained cytokine support and harsh immunosuppressive TME can still lead to the exhaustion and dysfunction of CAR-T cells. Biomaterial strategies such as hydrogels, toroidal-spiral particles, implantable biomaterials have been explored to enfold CAR-T cells and immunostimulatory substances, which can greatly enhance the retention and bioactivity of CAR-T cells [134,135,136,137] (Fig. 4).

Fig. 4
figure 4

Adjunctive delivery strategies of CAR-T cells in preclinical. The biomaterials, such as hydrogel, microneedles, and toroidal-spiral particles, can load CAR-T cells for scattered seeding in solid tumors, contributing to the improvement of CAR-T therapies. A CAR-T cells are wrapped in a special hydrogel, and the hydrogel will continuously release activated CAR-T cells at the site of the solid tumor. B CAR-T cells are loaded in the porous structure of microneedles. The needles will release CAR-T cells to kill tumor cells after puncture into tumor tissue. C TSP with inner toroidal-spiral channels facilitates CAR-T cell encapsulation, cytokine co-envelope near the surface for controlled release, to stimulate proliferation and activation of CAR-T cells. CAR-T cells are expanded and activated in the device and actively climb out of the collagen matrix toward the tumor cells after peritumoral implantation of the TSPs near the solid tumor

Hydrogel has recently been designed for the local delivery of CAR-T cells to treat solid tumors. Polymer-nanoparticle (PNP) hydrogels compose of water, cellulose polymers found in plants, and biodegradable nanoparticles. The tight mesh structure of hydrogels can load cytokines and CAR-T cells, which forms an enclosed and immune stimulatory environment for CAR-T cells. Post injection of the hydrogel complex through a needle, well-activated CAR-T cells can be slowly released as the hydrogel degrade and distribute in the tumor site. Controlled release of cytokines can support the long-term activation and persistence of CAR-T cells, thus augmenting CAR-T anti-tumor efficacy. Grosskopf found that mice injected with hydrogels containing CAR-T cells and cytokines had better efficacy compared to intravenous injections. Furthermore, the hydrogel significantly degraded in vivo in a few weeks and did not cause any unfavorable inflammatory reactions in the animals [138]. Zhou et al. designed an injectable CAR-T cell local delivery system based on the photo-crosslinked gelatin methacryloyl (GelMA) hydrogels. GelMA hydrogels can not only maintain good solubility but also form a three-dimensional structure by ultraviolet irradiation. It can support the survival and proliferation of CAR-T cells in the TME. GelMA hydrogels also can extend the retention time of CAR-T cells in the tumor site and gradually release them to eliminate tumor cells [139]. Compared to systemically delivered CAR-T cells, hydrogel-based CAR-T cells exhibit higher viability, proliferation, persistence, and anticancer activity. This approach may also prevent the harmful side effects of systemically administered CAR-T cells. These injectable hydrogels may be further developed in the future to allow for more precise regulation of CAR-T cells for long-term treatment lans [140, 141].

Transdermal delivery devices, a minimal and transdermal invasive to deliver drugs by the microneedle patch, can eliminate the possibility of tissue trauma and infection risk associated with injections. Transdermal delivery device makes it possible to conduct a prolonged release of a series of small molecular medications such as galanthamine, insulin, and antibodies [142, 143]. The first cryo-microneedles that could load live cells were created by Xu et al. The therapeutic cells can be delivered to the layer of immune cell-rich epidermis through the microneedles on the skin and can hold a superior persistence and activation. In mice, cells delivered by the cryo-microneedles retained viability and proliferative capability [144]. The depth and distribution of immune cells can be precisely controlled by adjusting the length and cell loading of the microneedles. The loaded cells are successfully delivered by pressing microneedles into the skin, and cryo-microneedle delivery keeps the loaded cells active for a long period. Gu and Li et al. construct a polymeric porous microneedle (PMN) patch to load CAR-T cells.The patch can be implanted in the tumor bed or in the post-surgical resection cavity to delivery CAR-T cells [145]. The microneedle patch offers a multipoint, scattered delivery strategy for CAR-T cells, which can enhance the CAR-T cells infiltration by overcoming physical barriers in solid tumors. More than half of the PMN loaded CAR-T cells were delivered to the tumor within 15 min according to their evaluation of the anticancer effects of CAR-T cells. The investigators also compared the intratumoral distribution of CAR-T cells through intratumoral injection and PMN-mediated delivery in the mice model with WM115 melanoma tumor. Comparing with intratumoral infusion, CAR-T cells delivered via PMN showed more prominent tumor infiltration. Collectively, transdermal administration systems based on microneedles offer a highly modular and efficient approach for CAR-T cell therapy.

Liu et al. designed a biodegradable and biocompatible Toroidal spiral particles (TSP) delivery platform that is universal for different types of lymphocytes. It has strength in high-capacity cell loading, programmable release, high efficacy, low toxicity, and minimally invasive operation. TSP can precisely control the delivery speed of cells, enable in-situ and local delivery of CAR-T cells. The team successfully loaded the MSLN-targeted CAR-T cells into the TSP platform, it triggers an immune response around the tumor and enhances the overall effect of the treatment. Compared to systemic and intratumoral injection, peritumoral delivery of MSLN CAR-T cells using the TSPs resulted in a superior antitumor effect [137].

The application of biomaterials in the adjunctive delivery of CAR-T cells provides a new idea for the treatment of solid tumors. In addition to the three adjunctive delivery strategies mentioned above, an increasing number of materials have been developed such as nitinol thin films, and the alginate scaffold, to enhance the viability, proliferation, persistence, and anti-cancer efficacies of CAR-T cells. Moreover, the CAR-T cells can spread from their implantation sites and circulate to kill distant tumor [146,147,148].

Conclusion

CAR-T cell therapy has already changed the therapeutic landscape of hematological malignancies. Enlightened by the extensive preclinical investigation and clinical experience of CAR-T therapy, clinical administration pattern of CAR-T cells plays a critical role in follow-up clinical response and side effect condition. Therefore, a comprehensive knowledge in current infusion dose scheme and delivery strategy of CAR-T cells is of necessity to guide the further therapeutic breakthrough and controllability. Facilitated by the advancement of multi-disciplinary technologies, novel regional CAR-T delivery strategy and biomaterial-based CAR-T delivery methods have been developed to treat refractory solid tumors. The ground-breaking outcomes have confirmed the significance and potential of these innovations, encouraging investigators and clinicians to make effort not only in CAR-T per se, but also in comprehensive consideration of technical combination an clinical practice.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

ADGRE2:

Adhesion G protein-coupled receptor E2

AGRE2:

Adhesion G protein-coupled receptor E2 inhibitors

AZA:

Azacytidine

AML:

Acute myeloid leukemia

BAFF:

B-cell activating factor of the TNF family

BCMA:

B cell maturation antigen

B-ALL:

B cell-acute lymphoblastic leukemia

BPDCN:

Blastic plasmacytoid dendritic cell neoplasm

CAR:

Chimeric antigen receptor

CAIX:

Carbonic anhydrase

CD44V6:

CD44 variant domain 6

CEA:

Carcinoembryonic antigen

CLL1:

C-type lectin-like molecule-1

C-MET:

Cellular-mesenchymal epithelial transition factor

CNS:

Central nervous system

CR:

Complete remissions

CRS:

Cytokine release syndrome

CT:

Computed tomography

Cy:

Cyclophosphamide

DLBCL:

Diffuse large B cell lymphoma

ECM:

Extracellular matrix

EGFR:

Epidermal growth factor receptor

EBV:

Epstein-barr virus

EPCAM:

Epithelial cell adhesion molecule

FLT3:

FMS-like tyrosine kinase

EMA:

European Medicines Agency

FDA:

American Food and Drug Administration

FL:

Follicular lymphoma

Flu:

Fludarabine

GD2:

Disialoganglioside

GFRα4:

GDNF Family Receptor α 4

GPC3:

Glypican-3

GPRC5D:

G protein-coupled receptor, class C, group 5, member D

GelMA:

Gelatin methacryloyl

HER2:

Human epidermal growth factor receptor 2

ICAM-1:

Intercellular cell adhesion molecule-1

ICANS:

Immune effector cell-associated neurotoxicity syndrome

KK-LC-1:

Kita-Kyushu lung cancer antigen-1

LEY:

Lewis y tetrasaccharide

LGR5:

Leucine-rich repeat-containing G-protein coupled receptor 5

MHLW:

Ministry of Health, Labour and Welfare

MSLN:

Mesothelin

MUC-1:

mucin 1

MM:

Multiple myeloma

MTD:

Maximum tolerated dose

NKG2D:

Natural killer group 2, member D

N.A:

Not available

NHL:

Non-hodgkin lymphoma

NMPA:

National Medical Products Administration

ORR:

Objective response rate

PD-L1:

Programmed cell death 1 ligand 1

PSCA:

Prostate stem cell antigen

PSMA:

Prostate-specific membrane antigen

PCL:

Plasma cell leukemia

PMN:

Porous microneedle

PNP:

Polymer-nanoparticle

ROR2:

Receptor tyrosine kinase like orphan receptor 2 Gene

r/r B-ALL:

Relapsed or refractory B cell precursor acute lymphoblastic leukemia

r/r BCL:

Relapsed and refractory B Cell Lymphoma

r/r LBCL:

Relapsed or refractory large B cell lymphoma

r/r MM:

Relapsed and refractory multiple myeloma

r/r MCL:

Relapsed and refractory mantle cell lymphoma

r/rAML:

Relapsed and refractory acute myeloid leukemia

r/rNHL:

Relapsed and refractory non-hodgkin lymphoma

SLAMF7:

Signaling lymphocytes activating molecule factor 7

TM4FS1:

Transmembrane 4 L six family 1

TRBC1:

T cell receptor beta constant 1

T-ALL:

T-cell acute lymphoblastic leukemia

TME:

Tumor microenvironment

T-NHL:

T cell non-hodgkin lymphoma

TSP:

Toroidal spiral particles

VEGFR2:

Vascular endothelial growth factor receptor 2

References

  1. Xiao X, Huang S, Chen S, Wang Y, Sun Q, Xu X, et al. Mechanisms of cytokine release syndrome and neurotoxicity of CAR T-cell therapy and associated prevention and management strategies. J Experimental Clin cancer Research: CR. 2021;40(1):367.

    Article  CAS  PubMed Central  Google Scholar 

  2. Anagnostou T, Brody JD. In CAR T cell-treated lymphomas, the T cell rich get richer. Nat Med. 2022;28(9):1757–8.

    Article  CAS  PubMed  Google Scholar 

  3. Lu J, Jiang G. The journey of CAR-T therapy in hematological malignancies. Mol Cancer. 2022;21(1):194.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Minson A, Hamad N, Cheah CY, Tam C, Blombery P, Westerman D, et al. CAR T cells and time-limited ibrutinib as treatment for relapsed/refractory mantle cell lymphoma: the phase 2 TARMAC study. Blood. 2024;143(8):673–84.

    Article  CAS  PubMed  Google Scholar 

  5. Kausar MA, Anwar S, El-Horany HE, Khan FH, Tyagi N, Najm MZ et al. Journey of CAR T–cells: emphasising the concepts and advancements in breast cancer (review). Int J Oncol. 2023;63(6).

  6. Cappell KM, Kochenderfer JN. Long-term outcomes following CAR T cell therapy: what we know so far. Nat Reviews Clin Oncol. 2023;20(6):359–71.

    Article  CAS  Google Scholar 

  7. Fitzgerald JC, Weiss SL, Maude SL, Barrett DM, Lacey SF, Melenhorst JJ, et al. Cytokine release syndrome after Chimeric Antigen Receptor T Cell Therapy for Acute Lymphoblastic Leukemia. Crit Care Med. 2017;45(2):e124–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Jin Z, Xiang R, Qing K, Li X, Zhang Y, Wang L, et al. The severe cytokine release syndrome in phase I trials of CD19-CAR-T cell therapy: a systematic review. Ann Hematol. 2018;97(8):1327–35.

    Article  CAS  PubMed  Google Scholar 

  9. Wei J, Liu Y, Wang C, Zhang Y, Tong C, Dai G, et al. The model of cytokine release syndrome in CAR T-cell treatment for B-cell non-hodgkin lymphoma. Signal Transduct Target Therapy. 2020;5(1):134.

    Article  CAS  Google Scholar 

  10. Roddie C, Dias J, O’Reilly MA, Abbasian M, Cadinanos-Garai A, Vispute K, et al. Durable responses and low toxicity after fast off-rate CD19 chimeric Antigen Receptor-T therapy in adults with relapsed or refractory B-Cell Acute Lymphoblastic Leukemia. J Clin Oncology: Official J Am Soc Clin Oncol. 2021;39(30):3352–63.

    Article  CAS  Google Scholar 

  11. Maalej KM, Merhi M, Inchakalody VP, Mestiri S, Alam M, Maccalli C, et al. CAR-cell therapy in the era of solid tumor treatment: current challenges and emerging therapeutic advances. Mol Cancer. 2023;22(1):20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Williams AD, Payne KK, Posey AD Jr., Hill C, Conejo-Garcia J, June CH et al. Immunotherapy for breast Cancer: current and future strategies. Curr Surg Rep. 2017;5.

  13. Brown CE, Alizadeh D, Starr R, Weng L, Wagner JR, Naranjo A, et al. Regression of Glioblastoma after chimeric Antigen receptor T-Cell therapy. N Engl J Med. 2016;375(26):2561–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Simula L, Fumagalli M, Vimeux L, Rajnpreht I, Icard P, Birsen G, et al. Mitochondrial metabolism sustains CD8(+) T cell migration for an efficient infiltration into solid tumors. Nat Commun. 2024;15(1):2203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Lv J, Zhao R, Wu D, Zheng D, Wu Z, Shi J, et al. Mesothelin is a target of chimeric antigen receptor T cells for treating gastric cancer. J Hematol Oncol. 2019;12(1):18.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Katz SC, Point GR, Cunetta M, Thorn M, Guha P, Espat NJ, et al. Regional CAR-T cell infusions for peritoneal carcinomatosis are superior to systemic delivery. Cancer Gene Ther. 2016;23(5):142–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Ma Q, He X, Zhang B, Guo F, Ou X, Yang Q, et al. A PD-L1-targeting chimeric switch receptor enhances efficacy of CAR-T cell for pleural and peritoneal metastasis. Signal Transduct Target Therapy. 2022;7(1):380.

    Article  Google Scholar 

  18. Brown CE, Badie B, Barish ME, Weng L, Ostberg JR, Chang WC, et al. Bioactivity and Safety of IL13Rα2-Redirected chimeric Antigen receptor CD8 + T cells in patients with recurrent glioblastoma. Clin cancer Research: Official J Am Association Cancer Res. 2015;21(18):4062–72.

    Article  CAS  Google Scholar 

  19. Mackensen A, Haanen J, Koenecke C, Alsdorf W, Wagner-Drouet E, Borchmann P, et al. CLDN6-specific CAR-T cells plus amplifying RNA vaccine in relapsed or refractory solid tumors: the phase 1 BNT211-01 trial. Nat Med. 2023;29(11):2844–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Qi C, Gong J, Li J, Liu D, Qin Y, Ge S, et al. Claudin18.2-specific CAR T cells in gastrointestinal cancers: phase 1 trial interim results. Nat Med. 2022;28(6):1189–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Adusumilli PS, Cherkassky L, Villena-Vargas J, Colovos C, Servais E, Plotkin J, et al. Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunity. Sci Transl Med. 2014;6(261):261ra151.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Priceman SJ, Tilakawardane D, Jeang B, Aguilar B, Murad JP, Park AK, et al. Regional Delivery of chimeric Antigen receptor-Engineered T cells effectively targets HER2(+) breast Cancer metastasis to the brain. Clin cancer Research: Official J Am Association Cancer Res. 2018;24(1):95–105.

    Article  CAS  Google Scholar 

  23. Niu H, Zhao P, Sun W. Biomaterials for chimeric antigen receptor T cell engineering. Acta Biomater. 2023;166:1–13.

    Article  CAS  PubMed  Google Scholar 

  24. Wagner DL, Fritsche E, Pulsipher MA, Ahmed N, Hamieh M, Hegde M, et al. Immunogenicity of CAR T cells in cancer therapy. Nat Reviews Clin Oncol. 2021;18(6):379–93.

    Article  Google Scholar 

  25. Han Z, Ma X, Ma G. Improving cell reinfusion to enhance the efficacy of chimeric antigen receptor T-cell therapy and alleviate complications. Heliyon. 2024;10(7):e28098.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Porter DL, Hwang WT, Frey NV, Lacey SF, Shaw PA, Loren AW, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med. 2015;7(303):303ra139.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Amini L, Silbert SK, Maude SL, Nastoupil LJ, Ramos CA, Brentjens RJ, et al. Preparing for CAR T cell therapy: patient selection, bridging therapies and lymphodepletion. Nat Reviews Clin Oncol. 2022;19(5):342–55.

    Article  Google Scholar 

  28. Jiang H, Li C, Yin P, Guo T, Liu L, Xia L, et al. Anti-CD19 chimeric antigen receptor-modified T-cell therapy bridging to allogeneic hematopoietic stem cell transplantation for relapsed/refractory B-cell acute lymphoblastic leukemia: an open-label pragmatic clinical trial. Am J Hematol. 2019;94(10):1113–22.

    Article  CAS  PubMed  Google Scholar 

  29. Feins S, Kong W, Williams EF, Milone MC, Fraietta JA. An introduction to chimeric antigen receptor (CAR) T-cell immunotherapy for human cancer. Am J Hematol. 2019;94(S1):S3–9.

    Article  CAS  PubMed  Google Scholar 

  30. Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med. 2013;368(16):1509–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371(16):1507–17.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Sheih A, Voillet V, Hanafi LA, DeBerg HA, Yajima M, Hawkins R, et al. Clonal kinetics and single-cell transcriptional profiling of CAR-T cells in patients undergoing CD19 CAR-T immunotherapy. Nat Commun. 2020;11(1):219.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Chaudhury A, Zhu X, Chu L, Goliaei A, June CH, Kearns JD, et al. Chimeric Antigen Receptor T Cell Therapies: a review of Cellular Kinetic-Pharmacodynamic modeling approaches. J Clin Pharmacol. 2020;60(Suppl 1):S147–59.

    CAS  PubMed  Google Scholar 

  34. Fraietta JA, Nobles CL, Sammons MA, Lundh S, Carty SA, Reich TJ, et al. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature. 2018;558(7709):307–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Brentjens RJ, Rivière I, Park JH, Davila ML, Wang X, Stefanski J, et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood. 2011;118(18):4817–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ling Y, Xuan L, Xu N, Huang F, Fan Z, Guo Z, et al. Busulfan Plus Fludarabine compared with Busulfan Plus Cyclophosphamide for AML undergoing HLA-Haploidentical hematopoietic cell transplantation: a Multicenter Randomized Phase III Trial. J Clin Oncology: Official J Am Soc Clin Oncol. 2023;41(29):4632–42.

    Article  CAS  Google Scholar 

  37. Hay KA, Hanafi LA, Li D, Gust J, Liles WC, Wurfel MM, et al. Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor-modified T-cell therapy. Blood. 2017;130(21):2295–306.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Rotte A, Frigault MJ, Ansari A, Gliner B, Heery C, Shah B. Dose-response correlation for CAR-T cells: a systematic review of clinical studies. J Immunother Cancer. 2022;10(12).

  39. Munshi NC, Anderson LD Jr., Shah N, Madduri D, Berdeja J, Lonial S, et al. Idecabtagene Vicleucel in Relapsed and Refractory multiple myeloma. N Engl J Med. 2021;384(8):705–16.

    Article  CAS  PubMed  Google Scholar 

  40. Zhao WH, Liu J, Wang BY, Chen YX, Cao XM, Yang Y, et al. A phase 1, open-label study of LCAR-B38M, a chimeric antigen receptor T cell therapy directed against B cell maturation antigen, in patients with relapsed or refractory multiple myeloma. J Hematol Oncol. 2018;11(1):141.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Fowler NH, Dickinson M, Dreyling M, Martinez-Lopez J, Kolstad A, Butler J, et al. Tisagenlecleucel in adult relapsed or refractory follicular lymphoma: the phase 2 ELARA trial. Nat Med. 2022;28(2):325–32.

    Article  CAS  PubMed  Google Scholar 

  42. Qu X, An G, Sui W, Wang T, Zhang X, Yang J et al. Phase 1 study of C-CAR088, a novel humanized anti-BCMA CAR T-cell therapy in relapsed/refractory multiple myeloma. J Immunother Cancer. 2022;10(9).

  43. Frey NV, Shaw PA, Hexner EO, Pequignot E, Gill S, Luger SM, et al. Optimizing chimeric Antigen receptor T-Cell therapy for adults with Acute Lymphoblastic Leukemia. J Clin Oncology: Official J Am Soc Clin Oncol. 2020;38(5):415–22.

    Article  CAS  Google Scholar 

  44. Morris EC, Neelapu SS, Giavridis T, Sadelain M. Cytokine release syndrome and associated neurotoxicity in cancer immunotherapy. Nat Rev Immunol. 2022;22(2):85–96.

    Article  CAS  PubMed  Google Scholar 

  45. Pishko A, Nasta SD. The role of novel immunotherapies in non-hodgkin lymphoma. Translational cancer Res. 2017;6(1):93–103.

    Article  CAS  Google Scholar 

  46. Foster MC, Savoldo B, Lau W, Rubinos C, Grover N, Armistead P, et al. Utility of a safety switch to abrogate CD19.CAR T-cell-associated neurotoxicity. Blood. 2021;137(23):3306–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Del Bufalo F, De Angelis B, Caruana I, Del Baldo G, De Ioris MA, Serra A, et al. GD2-CART01 for relapsed or Refractory High-Risk Neuroblastoma. N Engl J Med. 2023;388(14):1284–95.

    Article  PubMed  Google Scholar 

  48. Flugel CL, Majzner RG, Krenciute G, Dotti G, Riddell SR, Wagner DL, et al. Overcoming on-target, off-tumour toxicity of CAR T cell therapy for solid tumours. Nat Reviews Clin Oncol. 2023;20(1):49–62.

    Article  CAS  Google Scholar 

  49. Zheng Y, Nandakumar KS, Cheng K. Optimization of CAR-T cell-based therapies using small-molecule-based safety switches. J Med Chem. 2021;64(14):9577–91.

    Article  CAS  PubMed  Google Scholar 

  50. Cappell KM, Kochenderfer JN. A comparison of chimeric antigen receptors containing CD28 versus 4-1BB costimulatory domains. Nat Reviews Clin Oncol. 2021;18(11):715–27.

    Article  CAS  Google Scholar 

  51. Wang M, Munoz J, Goy A, Locke FL, Jacobson CA, Hill BT, et al. KTE-X19 CAR T-Cell therapy in relapsed or Refractory Mantle-Cell Lymphoma. N Engl J Med. 2020;382(14):1331–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Bishop MR, Dickinson M, Purtill D, Barba P, Santoro A, Hamad N, et al. Second-line tisagenlecleucel or Standard Care in Aggressive B-Cell Lymphoma. N Engl J Med. 2022;386(7):629–39.

    Article  CAS  PubMed  Google Scholar 

  53. Ali SA, Shi V, Maric I, Wang M, Stroncek DF, Rose JJ, et al. T cells expressing an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of multiple myeloma. Blood. 2016;128(13):1688–700.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson CA, et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory large B-Cell lymphoma. N Engl J Med. 2017;377(26):2531–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Shah NN, Johnson BD, Schneider D, Zhu F, Szabo A, Keever-Taylor CA, et al. Bispecific anti-CD20, anti-CD19 CAR T cells for relapsed B cell malignancies: a phase 1 dose escalation and expansion trial. Nat Med. 2020;26(10):1569–75.

    Article  CAS  PubMed  Google Scholar 

  56. Shah BD, Bishop MR, Oluwole OO, Logan AC, Baer MR, Donnellan WB, et al. KTE-X19 anti-CD19 CAR T-cell therapy in adult relapsed/refractory acute lymphoblastic leukemia: ZUMA-3 phase 1 results. Blood. 2021;138(1):11–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Neelapu SS, Dickinson M, Munoz J, Ulrickson ML, Thieblemont C, Oluwole OO, et al. Axicabtagene ciloleucel as first-line therapy in high-risk large B-cell lymphoma: the phase 2 ZUMA-12 trial. Nat Med. 2022;28(4):735–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Turtle CJ, Hanafi LA, Berger C, Hudecek M, Pender B, Robinson E, et al. Immunotherapy of non-hodgkin’s lymphoma with a defined ratio of CD8 + and CD4 + CD19-specific chimeric antigen receptor-modified T cells. Sci Transl Med. 2016;8(355):355ra116.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Locke FL, Ghobadi A, Jacobson CA, Miklos DB, Lekakis LJ, Oluwole OO, et al. Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): a single-arm, multicentre, phase 1–2 trial. Lancet Oncol. 2019;20(1):31–42.

    Article  CAS  PubMed  Google Scholar 

  60. Kamdar M, Solomon SR, Arnason J, Johnston PB, Glass B, Bachanova V, et al. Lisocabtagene maraleucel versus standard of care with salvage chemotherapy followed by autologous stem cell transplantation as second-line treatment in patients with relapsed or refractory large B-cell lymphoma (TRANSFORM): results from an interim analysis of an open-label, randomised, phase 3 trial. Lancet (London England). 2022;399(10343):2294–308.

    Article  CAS  PubMed  Google Scholar 

  61. Lin Y, Raje NS, Berdeja JG, Siegel DS, Jagannath S, Madduri D, et al. Idecabtagene vicleucel for relapsed and refractory multiple myeloma: post hoc 18-month follow-up of a phase 1 trial. Nat Med. 2023;29(9):2286–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Mailankody S, Matous JV, Chhabra S, Liedtke M, Sidana S, Oluwole OO, et al. Allogeneic BCMA-targeting CAR T cells in relapsed/refractory multiple myeloma: phase 1 UNIVERSAL trial interim results. Nat Med. 2023;29(2):422–9.

    Article  CAS  PubMed  Google Scholar 

  63. Raje N, Berdeja J, Lin Y, Siegel D, Jagannath S, Madduri D, et al. Anti-BCMA CAR T-Cell therapy bb2121 in relapsed or refractory multiple myeloma. N Engl J Med. 2019;380(18):1726–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Brudno JN, Maric I, Hartman SD, Rose JJ, Wang M, Lam N, et al. T cells genetically modified to Express an Anti-B-Cell Maturation Antigen Chimeric Antigen Receptor cause remissions of poor-prognosis relapsed multiple myeloma. J Clin Oncology: Official J Am Soc Clin Oncol. 2018;36(22):2267–80.

    Article  CAS  Google Scholar 

  65. Yee C, Thompson JA, Byrd D, Riddell SR, Roche P, Celis E, et al. Adoptive T cell therapy using antigen-specific CD8 + T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells. Proc Natl Acad Sci USA. 2002;99(25):16168–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Hu Y, Zhou Y, Zhang M, Zhao H, Wei G, Ge W, et al. Genetically modified CD7-targeting allogeneic CAR-T cell therapy with enhanced efficacy for relapsed/refractory CD7-positive hematological malignancies: a phase I clinical study. Cell Res. 2022;32(11):995–1007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Liu J, Zhang Y, Guo R, Zhao Y, Sun R, Guo S, et al. Targeted CD7 CAR T-cells for treatment of T-Lymphocyte leukemia and lymphoma and acute myeloid leukemia: recent advances. Front Immunol. 2023;14:1170968.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Guo J, He S, Zhu Y, Yu W, Yang D, Zhao X. Humanized CD30-Targeted Chimeric Antigen Receptor T Cells Exhibit Potent Preclinical Activity against Hodgkin’s lymphoma cells. Front cell Dev Biology. 2021;9:775599.

    Article  Google Scholar 

  69. Tschernia NP, Heiling H, Deal AM, Cheng C, Babinec C, Gonzalez M et al. Patient-reported outcomes in CD30-directed CAR-T cells against relapsed/refractory CD30 + lymphomas. J Immunother Cancer. 2023;11(8).

  70. Ramos CA, Grover NS, Beaven AW, Lulla PD, Wu MF, Ivanova A, et al. Anti-CD30 CAR-T cell therapy in relapsed and refractory Hodgkin Lymphoma. J Clin Oncology: Official J Am Soc Clin Oncol. 2020;38(32):3794–804.

    Article  CAS  Google Scholar 

  71. Teachey DT, Lacey SF, Shaw PA, Melenhorst JJ, Maude SL, Frey N, et al. Identification of predictive biomarkers for Cytokine Release Syndrome after chimeric Antigen receptor T-cell therapy for Acute Lymphoblastic Leukemia. Cancer Discov. 2016;6(6):664–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Abramson JS, Palomba ML, Gordon LI, Lunning MA, Wang M, Arnason J, et al. Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study. Lancet (London England). 2020;396(10254):839–52.

    Article  PubMed  Google Scholar 

  73. Park JH, Rivière I, Gonen M, Wang X, Sénéchal B, Curran KJ, et al. Long-term follow-up of CD19 CAR therapy in Acute Lymphoblastic Leukemia. N Engl J Med. 2018;378(5):449–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, Bagg A, et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011;3(95):95ra73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Meader N, King K, Llewellyn A, Norman G, Brown J, Rodgers M, et al. A checklist designed to aid consistency and reproducibility of GRADE assessments: development and pilot validation. Syst Reviews. 2014;3:82.

    Article  Google Scholar 

  76. Xu J, Chen LJ, Yang SS, Sun Y, Wu W, Liu YF, et al. Exploratory trial of a biepitopic CAR T-targeting B cell maturation antigen in relapsed/refractory multiple myeloma. Proc Natl Acad Sci USA. 2019;116(19):9543–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Geyer MB, Rivière I, Sénéchal B, Wang X, Wang Y, Purdon TJ, et al. Autologous CD19-Targeted CAR T cells in patients with residual CLL following initial Purine Analog-based therapy. Mol Therapy: J Am Soc Gene Therapy. 2018;26(8):1896–905.

    Article  CAS  Google Scholar 

  78. Zhang Q, Hu H, Chen SY, Liu CJ, Hu FF, Yu J, et al. Transcriptome and Regulatory Network Analyses of CD19-CAR-T immunotherapy for B-ALL. Genom Proteom Bioinform. 2019;17(2):190–200.

    Article  CAS  Google Scholar 

  79. Su T, Ying Z, Lu XA, He T, Song Y, Wang X, et al. The clinical outcomes of fresh versus cryopreserved CD19-directed chimeric antigen receptor T cells in non-hodgkin lymphoma patients. Cryobiology. 2020;96:106–13.

    Article  CAS  PubMed  Google Scholar 

  80. Maschan M, Caimi PF, Reese-Koc J, Sanchez GP, Sharma AA, Molostova O, et al. Multiple site place-of-care manufactured anti-CD19 CAR-T cells induce high remission rates in B-cell malignancy patients. Nat Commun. 2021;12(1):7200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Beck C, Casey NP, Persiconi I, Moharrami NN, Sike A, Jin Y et al. Development of a TGFβ-IL-2/15 switch receptor for Use in Adoptive Cell Therapy. Biomedicines. 2023;11(2).

  82. Xu H, Cao W, Huang L, Xiao M, Cao Y, Zhao L, et al. Effects of cryopreservation on chimeric antigen receptor T cell functions. Cryobiology. 2018;83:40–7.

    Article  CAS  PubMed  Google Scholar 

  83. Panch SR, Srivastava SK, Elavia N, McManus A, Liu S, Jin P, et al. Effect of cryopreservation on autologous chimeric Antigen receptor T cell characteristics. Mol Therapy: J Am Soc Gene Therapy. 2019;27(7):1275–85.

    Article  CAS  Google Scholar 

  84. Dreyzin A, Panch SR, Shalabi H, Yates B, Highfill SL, Jin P, et al. Cryopreserved anti-CD22 and bispecific anti-CD19/22 CAR T cells are as effective as freshly infused cells. Mol Therapy Methods Clin Dev. 2023;28:51–61.

    Article  CAS  Google Scholar 

  85. Brezinger-Dayan K, Itzhaki O, Melnichenko J, Kubi A, Zeltzer LA, Jacoby E, et al. Impact of cryopreservation on CAR T production and clinical response. Front Oncol. 2022;12:1024362.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Hanley PJ. Fresh versus frozen: effects of Cryopreservation on CAR T cells. Mol Therapy: J Am Soc Gene Therapy. 2019;27(7):1213–4.

    Article  CAS  Google Scholar 

  87. Lee SY, Olsen P, Lee DH, Kenoyer AL, Budde LE, O’Steen S et al. Preclinical Optimization of a CD20-specific Chimeric Antigen Receptor Vector and Culture Conditions. Journal of immunotherapy (Hagerstown, Md: 1997). 2018;41(1):19–31.

  88. Hughes SM, Shu Z, Levy CN, Ferre AL, Hartig H, Fang C, et al. Cryopreservation of Human Mucosal leukocytes. PLoS ONE. 2016;11(5):e0156293.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Liu G, Rui W, Zhao X, Lin X. Enhancing CAR-T cell efficacy in solid tumors by targeting the tumor microenvironment. Cell Mol Immunol. 2021;18(5):1085–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Theruvath J, Sotillo E, Mount CW, Graef CM, Delaidelli A, Heitzeneder S, et al. Locoregionally administered B7-H3-targeted CAR T cells for treatment of atypical teratoid/rhabdoid tumors. Nat Med. 2020;26(5):712–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Tsai CH, Singh AP, Xia CQ, Wang H. Development of minimal physiologically-based pharmacokinetic-pharmacodynamic models for characterizing cellular kinetics of CAR T cells following local deliveries in mice. J Pharmacokinet Pharmacodyn. 2022;49(5):525–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Fousek K, Ahmed N. The evolution of T-cell therapies for solid malignancies. Clin cancer Research: Official J Am Association Cancer Res. 2015;21(15):3384–92.

    Article  CAS  Google Scholar 

  93. Melero I, Castanon E, Alvarez M, Champiat S, Marabelle A. Intratumoural administration and tumour tissue targeting of cancer immunotherapies. Nat Reviews Clin Oncol. 2021;18(9):558–76.

    Article  CAS  Google Scholar 

  94. Sridhar P, Petrocca F. Regional Delivery of Chimeric Antigen Receptor (CAR) T-Cells for Cancer Therapy. Cancers. 2017;9(7).

  95. Qin YT, Li YP, He XW, Wang X, Li WY, Zhang YK. Biomaterials promote in vivo generation and immunotherapy of CAR-T cells. Front Immunol. 2023;14:1165576.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Lanitis E, Dangaj D, Irving M, Coukos G. Mechanisms regulating T-cell infiltration and activity in solid tumors. Annals Oncology: Official J Eur Soc Med Oncol. 2017;28(suppl12):xii18–32.

    Article  CAS  Google Scholar 

  97. Dong H, Xu X, Wang L, Mo R. Advances in living cell-based anticancer therapeutics. Biomaterials Sci. 2020;8(9):2344–65.

    Article  CAS  Google Scholar 

  98. Albelda SM. CAR T cell therapy for patients with solid tumours: key lessons to learn and unlearn. Nat Reviews Clin Oncol. 2024;21(1):47–66.

    Article  Google Scholar 

  99. Cherkassky L, Hou Z, Amador-Molina A, Adusumilli PS. Regional CAR T cell therapy: an ignition key for systemic immunity in solid tumors. Cancer Cell. 2022;40(6):569–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Sagnella SM, White AL, Yeo D, Saxena P, van Zandwijk N, Rasko JEJ. Locoregional delivery of CAR-T cells in the clinic. Pharmacol Res. 2022;182:106329.

    Article  CAS  PubMed  Google Scholar 

  101. Shyr CR, Liu LC, Chien HS, Huang CP. Immunotherapeutic agents for Intratumoral Immunotherapy. Vaccines. 2023;11(11).

  102. Sheth RA, Murthy R, Hong DS, Patel S, Overman MJ, Diab A, et al. Assessment of Image-guided Intratumoral Delivery of Immunotherapeutics in patients with Cancer. JAMA Netw open. 2020;3(7):e207911.

    Article  PubMed  Google Scholar 

  103. Marabelle A, Andtbacka R, Harrington K, Melero I, Leidner R, de Baere T, et al. Starting the fight in the tumor: expert recommendations for the development of human intratumoral immunotherapy (HIT-IT). Annals Oncology: Official J Eur Soc Med Oncol. 2018;29(11):2163–74.

    Article  CAS  Google Scholar 

  104. Levy MJ, Alberts SR, Bamlet WR, Burch PA, Farnell MB, Gleeson FC, et al. EUS-guided fine-needle injection of gemcitabine for locally advanced and metastatic pancreatic cancer. Gastrointest Endosc. 2017;86(1):161–9.

    Article  PubMed  Google Scholar 

  105. Tang N, Cheng C, Zhang X, Qiao M, Li N, Mu W et al. TGF-β inhibition via CRISPR promotes the long-term efficacy of CAR T cells against solid tumors. JCI Insight. 2020;5(4).

  106. Tchou J, Zhao Y, Levine BL, Zhang PJ, Davis MM, Melenhorst JJ, et al. Safety and Efficacy of Intratumoral Injections of Chimeric Antigen Receptor (CAR) T cells in metastatic breast Cancer. Cancer Immunol Res. 2017;5(12):1152–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. 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;14(1):118.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Papa S, Adami A, Metoudi M, Beatson R, George MS, Achkova D et al. Intratumoral pan-ErbB targeted CAR-T for head and neck squamous cell carcinoma: interim analysis of the T4 immunotherapy study. J Immunother Cancer. 2023;11(6).

  109. Hong WX, Haebe S, Lee AS, Westphalen CB, Norton JA, Jiang W, et al. Intratumoral Immunotherapy for early-stage solid tumors. Clin cancer Research: Official J Am Association Cancer Res. 2020;26(13):3091–9.

    Article  CAS  Google Scholar 

  110. Saied A, Licata L, Burga RA, Thorn M, McCormack E, Stainken BF, et al. Neutrophil:lymphocyte ratios and serum cytokine changes after hepatic artery chimeric antigen receptor-modified T-cell infusions for liver metastases. Cancer Gene Ther. 2014;21(11):457–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Katz SC, Moody AE, Guha P, Hardaway JC, Prince E, LaPorte J et al. HITM-SURE: hepatic immunotherapy for metastases phase ib anti-CEA CAR-T study utilizing pressure enabled drug delivery. J Immunother Cancer. 2020;8(2).

  112. Katz SC, Burga RA, McCormack E, Wang LJ, Mooring W, Point GR, et al. Phase I hepatic immunotherapy for metastases study of intra-arterial chimeric Antigen receptor-modified T-cell therapy for CEA + liver metastases. Clin cancer Research: Official J Am Association Cancer Res. 2015;21(14):3149–59.

    Article  CAS  Google Scholar 

  113. Hege KM, Bergsland EK, Fisher GA, Nemunaitis JJ, Warren RS, McArthur JG, et al. Safety, tumor trafficking and immunogenicity of chimeric antigen receptor (CAR)-T cells specific for TAG-72 in colorectal cancer. J Immunother Cancer. 2017;5:22.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Katz SC, Hardaway J, Prince E, Guha P, Cunetta M, Moody A, et al. HITM-SIR: phase Ib trial of intraarterial chimeric antigen receptor T-cell therapy and selective internal radiation therapy for CEA(+) liver metastases. Cancer Gene Ther. 2020;27(5):341–55.

    Article  CAS  PubMed  Google Scholar 

  115. Esselen KM, Rodriguez N, Growdon W, Krasner C, Horowitz NS, Campos S. Patterns of recurrence in advanced epithelial ovarian, fallopian tube and peritoneal cancers treated with intraperitoneal chemotherapy. Gynecol Oncol. 2012;127(1):51–4.

    Article  CAS  PubMed  Google Scholar 

  116. Wang X, Zhou J, Wang Y, Zhu Z, Lu Y, Wei Y, et al. A phase I clinical and pharmacokinetic study of paclitaxel liposome infused in non-small cell lung cancer patients with malignant pleural effusions. Eur J cancer (Oxford England: 1990). 2010;46(8):1474–80.

    Article  CAS  Google Scholar 

  117. Danson SJ, Conner J, Edwards JG, Blyth KG, Fisher PM, Muthana M, et al. Oncolytic herpesvirus therapy for mesothelioma - A phase I/IIa trial of intrapleural administration of HSV1716. Lung cancer (Amsterdam. Netherlands). 2020;150:145–51.

    Google Scholar 

  118. Qian S, Chen J, Zhao Y, Zhu X, Dai D, Qin L et al. Intraperitoneal administration of carcinoembryonic antigen-directed chimeric antigen receptor T cells is a robust delivery route for effective treatment of peritoneal carcinomatosis from colorectal cancer in pre-clinical study. Cytotherapy. 2023.

  119. Ranoa DRE, Sharma P, Schane CP, Lewis AN, Valdez E, Marada V et al. Single CAR-T cell treatment controls disseminated ovarian cancer in a syngeneic mouse model. J Immunother Cancer. 2023;11(5).

  120. Adusumilli PS, Zauderer MG, Rivière I, Solomon SB, Rusch VW, O’Cearbhaill RE, et al. A phase I Trial of Regional Mesothelin-targeted CAR T-cell therapy in patients with malignant Pleural Disease, in combination with the Anti-PD-1 Agent Pembrolizumab. Cancer Discov. 2021;11(11):2748–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Lu Z, Wang J, Wientjes MG, Au JL. Intraperitoneal therapy for peritoneal cancer. Future Oncol (London England). 2010;6(10):1625–41.

    Article  Google Scholar 

  122. Castelletti L, Yeo D, van Zandwijk N, Rasko JEJ. Anti-mesothelin CAR T cell therapy for malignant mesothelioma. Biomark Res. 2021;9(1):11.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Deshet-Unger N, Horn G, Rawet-Slobodkin M, Waks T, Laskov I, Michaan N et al. Comparing intraperitoneal and intravenous personalized ErbB2CAR-T for the treatment of epithelial ovarian Cancer. Biomedicines. 2022;10(9).

  124. Hiltbrunner S, Britschgi C, Schuberth P, Bankel L, Nguyen-Kim TDL, Gulati P, et al. Local delivery of CAR T cells targeting fibroblast activation protein is safe in patients with pleural mesothelioma: first report of FAPME, a phase I clinical trial. Annals Oncology: Official J Eur Soc Med Oncol. 2021;32(1):120–1.

    Article  CAS  Google Scholar 

  125. Koneru M, O’Cearbhaill R, Pendharkar S, Spriggs DR, Brentjens RJ. A phase I clinical trial of adoptive T cell therapy using IL-12 secreting MUC-16(ecto) directed chimeric antigen receptors for recurrent ovarian cancer. J Translational Med. 2015;13:102.

    Article  Google Scholar 

  126. Donovan LK, Delaidelli A, Joseph SK, Bielamowicz K, Fousek K, Holgado BL, et al. Locoregional delivery of CAR T cells to the cerebrospinal fluid for treatment of metastatic medulloblastoma and ependymoma. Nat Med. 2020;26(5):720–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Brown CE, Rodriguez A, Palmer J, Ostberg JR, Naranjo A, Wagner JR, et al. Off-the-shelf, steroid-resistant, IL13Rα2-specific CAR T cells for treatment of glioblastoma. Neurooncology. 2022;24(8):1318–30.

    CAS  Google Scholar 

  128. Kim K, Gwak HS, Han N, Hong EK, Choi BK, Lee S, et al. Chimeric Antigen Receptor T cells with modified Interleukin-13 preferentially recognize IL13Rα2 and suppress malignant glioma: a preclinical study. Front Immunol. 2021;12:715000.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Vitanza NA, Ronsley R, Choe M, Henson C, Breedt M, Barrios-Anderson A, et al. Locoregional CAR T cells for children with CNS tumors: clinical procedure and catheter safety. Volume 36. New York, NY: Neoplasia; 2023. p. 100870.

    Google Scholar 

  130. Vitanza NA, Wilson AL, Huang W, Seidel K, Brown C, Gustafson JA, et al. Intraventricular B7-H3 CAR T cells for diffuse intrinsic pontine glioma: preliminary first-in-human bioactivity and safety. Cancer Discov. 2023;13(1):114–31.

    Article  CAS  PubMed  Google Scholar 

  131. Vitanza NA, Johnson AJ, Wilson AL, Brown C, Yokoyama JK, Künkele A, et al. Locoregional infusion of HER2-specific CAR T cells in children and young adults with recurrent or refractory CNS tumors: an interim analysis. Nat Med. 2021;27(9):1544–52.

    Article  CAS  PubMed  Google Scholar 

  132. Liu Z, Zhou J, Yang X, Liu Y, Zou C, Lv W, et al. Safety and antitumor activity of GD2-Specific 4SCAR-T cells in patients with glioblastoma. Mol Cancer. 2023;22(1):3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Waldmann TA, Lugli E, Roederer M, Perera LP, Smedley JV, Macallister RP, et al. Safety (toxicity), pharmacokinetics, immunogenicity, and impact on elements of the normal immune system of recombinant human IL-15 in rhesus macaques. Blood. 2011;117(18):4787–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Stephan SB, Taber AM, Jileaeva I, Pegues EP, Sentman CL, Stephan MT. Biopolymer implants enhance the efficacy of adoptive T-cell therapy. Nat Biotechnol. 2015;33(1):97–101.

    Article  CAS  PubMed  Google Scholar 

  135. Wang C, Ye Y, Hochu GM, Sadeghifar H, Gu Z. Enhanced Cancer Immunotherapy by Microneedle Patch-assisted delivery of Anti-PD1 antibody. Nano Lett. 2016;16(4):2334–40.

    Article  CAS  PubMed  Google Scholar 

  136. Liu Y, Xiao L, Joo KI, Hu B, Fang J, Wang P. In situ modulation of dendritic cells by injectable thermosensitive hydrogels for cancer vaccines in mice. Biomacromolecules. 2014;15(10):3836–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Tang H, Zaroudi M, Zhu Y, Cheng A, Qin L, Zhang B, et al. Toroidal-spiral particles as a CAR-T cell delivery device for solid tumor immunotherapy. J Controlled Release: Official J Controlled Release Soc. 2023;362:620–30.

    Article  CAS  Google Scholar 

  138. Grosskopf AK, Labanieh L, Klysz DD, Roth GA, Xu P, Adebowale O, et al. Delivery of CAR-T cells in a transient injectable stimulatory hydrogel niche improves treatment of solid tumors. Sci Adv. 2022;8(14):eabn8264.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Zhou W, Lei S, Liu M, Li D, Huang Y, Hu X, et al. Injectable and photocurable CAR-T cell formulation enhances the anti-tumor activity to melanoma in mice. Biomaterials. 2022;291:121872.

    Article  CAS  PubMed  Google Scholar 

  140. Riley RS, June CH, Langer R, Mitchell MJ. Delivery technologies for cancer immunotherapy. Nat Rev Drug Discovery. 2019;18(3):175–96.

    Article  CAS  PubMed  Google Scholar 

  141. Wang C, Wang J, Zhang X, Yu S, Wen D, Hu Q et al. In situ formed reactive oxygen species-responsive scaffold with gemcitabine and checkpoint inhibitor for combination therapy. Sci Transl Med. 2018;10(429).

  142. Ye Y, Yu J, Wen D, Kahkoska AR, Gu Z. Polymeric microneedles for transdermal protein delivery. Adv Drug Deliv Rev. 2018;127:106–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Cegla J. Microneedle-array patches loaded with hypoxia-sensitive vesicles provide fast glucose-responsive insulin delivery. Ann Clin Biochem. 2015;52(6):720.

    Article  PubMed  Google Scholar 

  144. Chang H, Chew SWT, Zheng M, Lio DCS, Wiraja C, Mei Y, et al. Cryomicroneedles for transdermal cell delivery. Nat Biomedical Eng. 2021;5(9):1008–18.

    Article  CAS  Google Scholar 

  145. Li H, Wang Z, Ogunnaike EA, Wu Q, Chen G, Hu Q, et al. Scattered seeding of CAR T cells in solid tumors augments anticancer efficacy. Natl Sci Rev. 2022;9(3):nwab172.

    Article  CAS  PubMed  Google Scholar 

  146. Coon ME, Stephan SB, Gupta V, Kealey CP, Stephan MT. Nitinol thin films functionalized with CAR-T cells for the treatment of solid tumours. Nat Biomedical Eng. 2020;4(2):195–206.

    Article  CAS  Google Scholar 

  147. Agarwalla P, Ogunnaike EA, Ahn S, Froehlich KA, Jansson A, Ligler FS, et al. Bioinstructive implantable scaffolds for rapid in vivo manufacture and release of CAR-T cells. Nat Biotechnol. 2022;40(8):1250–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Smith TT, Moffett HF, Stephan SB, Opel CF, Dumigan AG, Jiang X, et al. Biopolymers codelivering engineered T cells and STING agonists can eliminate heterogeneous tumors. J Clin Investig. 2017;127(6):2176–91.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The schematics were created with Biorender.com. Xinyu Gu and Yalan Zhang contributed equally to this work.

Funding

This work was supported by National Natural Science Foundation of China (81972878,82172733), the National Key Research and Development Program of China (2023YFC3403704), the Key Research and Development Program of Sichuan Province (2022ZDZX0024) and the International Cooperation Project of Sichuan Province (2021YFH0002).

Author information

Authors and Affiliations

Authors

Contributions

WW conceived and presented the article idea and supervised the whole work. XYG and YLZ collected the information, wrote and harmonized the manuscript. WLZ was a major contributor in designing the figures and editing the manuscript.FLW, FYY and HZG participated in collecting data and reviewing the manuscript.

Corresponding author

Correspondence to Wei Wang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gu, X., Zhang, Y., Zhou, W. et al. Infusion and delivery strategies to maximize the efficacy of CAR-T cell immunotherapy for cancers. Exp Hematol Oncol 13, 70 (2024). https://doi.org/10.1186/s40164-024-00542-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40164-024-00542-2

Keywords