Skip to main content
Molecular Therapy Oncolytics logoLink to Molecular Therapy Oncolytics
. 2021 Dec 21;24:194–204. doi: 10.1016/j.omto.2021.12.018

Expanding the role of interventional oncology for advancing precision immunotherapy of solid tumors

Yasushi Kimura 1, Mario Ghosn 2, Waseem Cheema 3, Prasad S Adusumilli 3, Stephen B Solomon 2, Govindarajan Srimathveeralli 1,4,5,
PMCID: PMC8752905  PMID: 35036524

Abstract

Adoptive cell therapy with chimeric antigen receptors (CAR) T cells has proven effective for hematologic malignancies, but success in solid tumors has been impeded by poor intratumoral infiltration, exhaustion of effector cells from antigen burden, and an immunosuppressive tumor microenvironment. Results from recent clinical trials and preclinical studies lend promising evidence of locoregional approaches for CAR T cell delivery, priming the tumor microenvironment, and performing adjuvant therapies that sustain T cell activity. Interventional oncology is a subspeciality of interventional radiology where imaging guidance is used to perform percutaneous and catheter-directed procedures for localized, non-surgical therapy or interrogation of solid tumors. Interventional oncology provides unique synergies with immunotherapy, which has been well-studied to improve treatment efficacy while reducing toxicities associated with systemic treatment. Besides aiding in CAR T cell delivery, priming, or the stimulation of the tumor microenvironment to promote effector survival and function, interventional oncology can also aid in the monitoring of treatment response through selective, multiplex tumor sampling and catheter-based venous sampling. This review presents an overview of interventional oncology, its various procedures, and its potential for advancing CAR T cell immunotherapy of solid tumors.

Keywords: CAR T cell therapy, interventional oncology, interventional radiology, locoregional cell delivery, percutaneous ablation, embolization, intraarterial drug delivery, image guided biopsy, venous sampling

Graphical abstract

graphic file with name fx1.jpg


Interventional oncology can advance adoptive cell therapy of solid tumors through locoregional approaches that improve biodistribution while reducing systemic toxicities. Interventional oncology techniques can prime the tumor microenvironment for chimeric antigen receptor T cell entry and function by aiding accurate monitoring of therapeutic outcomes by selective venous sampling or multifocal biopsy.

Introduction

T lymphocytes (T cells) play a central role in cancer immunotherapy. T cells are capable of long-term antigen-specific cytotoxicity after activation by antigen-presenting cells. However, cancer cells develop multiple pathways to evade, modify, and suppress the immune system despite ongoing immunosurveillance and anti-tumor activity.1, 2, 3 The development of immune checkpoint inhibitors (ICIs) has enabled efficacious immunotherapy of several types of cancer,4,5 yet clinical evidence suggests that only a fraction of patients demonstrate a durable or complete response to treatment, presenting an opportunity for improvement in both treatment efficacy and the patient population who can benefit from this therapy.6 Tumor-infiltrating lymphocytes (TILs) are a major determinant of ICI efficacy.7,8 Tumors with TILs (“hot tumors”) correlate with the expression of the PD-1/PD-L1 and are predictive of response to ICIs, while tumors without TILs (“cold tumors”) have a muted response.9, 10, 11 Recent research has led to the stratification of the tumor microenvironment (TME) into three major subtypes: immune-inflamed, immune-excluded, and immune-desert phenotypes.12 There is growing evidence that altering the TME by increasing TIL presence and function can improve cancer immunotherapy independent of disease characteristics or stage, with an increased emphasis on approaches that can yield this outcome.11,13

Adoptive cell therapy

Adoptive cell therapy (ACT) is an innovative approach used to increase the number of TILs wherein the patients’ own immune cells are collected, cultured ex vivo with or without additional genetic manipulation or biological modification, expanded, and reinfused into the patient. Immune cells introduced into the patient through this technique exhibit high tumor-antigen specificity and anti-cancer potency. However, three challenges emerged that limited the broader utility of ACT using TILs: TILs must be extracted from tumor samples, the proliferation and function of TILs is heavily dependent on age and gender;14 and the time between TIL extraction and administration often takes several weeks, all of which can be detrimental to patients with rapidly progressing cancers. These hurdles have impeded to the use of TILs in the broader clinical setting despite strong preliminary data in patients with several different cancers.15, 16, 17

Engineered T cell-based therapies

To overcome the drawbacks of ACT with TILs, two different approaches have been proposed using autologous genetically engineered immune cells, namely T cell receptor (TCR) and chimeric antigen receptor (CAR) T cell therapies. During TCR therapy, circulating T cells are collected and undergo TCR gene manipulation to augment recognition of the tumor antigen on the major histocompatibility complex (MHC).18 TCR has the potential limitation of treatment failure due to both antigen loss or MHC expression loss, whereas only antigen loss can affect the CAR T cells.19,20 CAR T cells represent an alternate non-MHC-dependent approach to cellular immunotherapy. The CAR comprises three sections, including an extracellular antigen recognition domain derived from monoclonal antibodies, the transmembrane domain, and the intracellular T cell activation domain.21 This approach has demonstrated success in the treatment of several hematologic malignancies in patients, such as acute lymphocytic leukemia (ALL) and diffuse large B cell lymphoma.22,23 CAR T cells targeting CD19 expressed in B cells have demonstrated very high objective response rates in patients with refractory ALL, with sustained responses in the majority of patients.

Current barriers to the effective CAR T treatment of solid tumors

While research to extend the application of CAR T cells to solid tumors is currently ongoing, there are challenges that have limited its efficacy when compared with hematologic malignancies. Obstacles include T cell localization, infiltration, systemic toxicity, insufficient activation due to immunosuppressive TME, and the lack of suitable tools for monitoring CAR T cell activity. Tumor-specific localization and infiltration have been identified as major impediments for the application of CAR T cell therapy in solid tumors (Figure 1A). Compared with hematological malignancies, CAR T cells need to home in to the tumor site and penetrate the tumor mass. Solid tumors secrete chemokines that can interfere with T cell localization and infiltration,24 which are further exacerbated by the TME having a disorganized extracellular matrix (ECM), immature or dysfunctional blood vessels, and high interstitial fluid pressure (IFP).25,26 Patients undergoing ICI and CAR T cell therapies can experience systemic toxicities due to intravenous treatment delivery.27,28 Clinical benefits and systemic toxicity are usually in conflict and may limit the maximum therapeutic dose a patient can receive. Immunosuppressive TME can impede CAR T cell activity in solid tumors (Figure 1B). The presence of other immune cells such as myeloid-derived suppressor cells, regulatory T cells (T regs), and tumor-associated macrophages (TAMs) can blunt CAR T cell effectiveness through multifactorial pathways.29 Monitoring the persistent activity of CAR T cells is also essential for evaluation of treatment response and making adjustments to the dosing regimen. Flow cytometry and local tumor biopsies have been used as tools for tracking CAR T cells.30 However, these approaches may provide skewed results, as blood samples from systemic circulation or tumor samples from a central core may not represent actual CAR T cell levels or the activity within heterogeneous solid tumors (Figure 1C).31

Figure 1.

Figure 1

Obstacles for CAR T cell application in solid tumors

Combination or adjuvant strategies that can alleviate these hurdles to augment efficacy of CAR T cells are therefore considered to be crucial for the success of solid tumor immunotherapy. Preclinical studies using CAR T cell therapy with adjuvant chemotherapy have demonstrated potential synergies through increased calreticulin and chemokine expression, augmented tumor antigen exposure,32,33 improved dendritic cell maturation and activation,34 and reduced autoimmunity to CAR T cells.35 Radiotherapy in combination with CAR T cells has also shown promise in preclinical studies. Radiotherapy is known to enhance MHC expression on tumor cells,36 induce chemokine release to help trafficking,37 and help dendritic cell maturation,38 factors that can aid CAR T cell infiltration and function. However, radiation can cause the death of T cells due to the inherent radiation sensitivity of lymphocytes.39,40 Finally, adjuvant use of ICIs can also improve CAR T cell therapy in solid tumors. Both preclinical studies and phase I clinical trials revealed that ICIs can improve CAR T cell activity.41, 42, 43, 44, 45, 46 While these combination therapy approaches are promising, barriers to effective treatment such as tumor heterogeneity, limited life span of functional CAR T cells, morbidity from multiple therapeutic agents, and on-target/off-tumor toxicity remain major challenges. We propose that interventional oncology provides a novel procedural approach to tackle several of these challenges.

Interventional oncology

Interventional oncology is a sub-discipline of interventional radiology (IR) that utilizes imaging guidance to perform minimally invasive, non-surgical procedures to treat patients with cancer.47, 48, 49 The interventions are performed using imaging modalities such as X-ray fluoroscopy, ultrasound (US), computed tomography (CT), and magnetic resonance imaging (MRI), which support intraprocedural imaging and prognostication of outcomes. Interventional oncology procedures can be broadly classified into two categories based on the approach used to gain access to the anatomical location of interest: (1) vascular procedures (V-IO - Vascular Interventional Oncology), where blood vessels or lymph ducts are used to navigate catheters to the tumor, and (2) percutaneous procedures (P-IO - Percutaneous Interventional Oncology), where cross-sectional imaging is used to guide percutaneous needle placement into the tumor.

Delivery approaches with interventional oncology

Vascular interventional oncology

During V-IO, a catheter is advanced under fluoroscopy to the primary artery feeding tumors, allowing for the selective and high-dose delivery of therapeutic material. Intraarterial injection of a cytotoxic drug or selective internal radiation therapy are image-guided approaches for the local delivery of chemotherapy/radiotherapy with the capacity to achieve very high local dosages.50, 51, 52 Embolization is a method of deliberately occluding a blood vessel with embolic materials. Several embolic materials are available for V-IO, such as gelatin sponges, microspheres, metallic coils, and liquid agents.53 The embolic materials can be classified as temporary (such as gelatin sponges) or permanent (such as metallic coils), and the choice of embolic material depends on the target vessel, location, and clinical objective. As for tumors, occluding the feeding arteries causes the tumor to become ischemic, infarcted, or necrotic, resulting in tumor shrinkage or a reduction of tumor growth.54, 55, 56, 57, 58, 59 In addition, embolization can be effective for tumors with active bleeding60, 61, 62 or a reduction of intraoperative blood loss.63, 64, 65, 66 The combined transcatheter delivery of both chemotherapy and embolization to hypervascular tumors is called transarterial chemoembolization (TACE). TACE has been the first-line treatment for hepatocellular carcinoma (HCC) patients with an intermediate stage (Barcelona Clinic Liver Cancer Stage B).51,55,67 As another type of V-IO treatment, selective internal radiation therapy (SIRT) is a developing modality for the treatment of patients with unresectable liver tumors such as HCC, colorectal liver metastasis, and neuroendocrine liver metastases (Figure 2A).68, 69, 70 The procedure consists of a transcatheter injection of radioactive particles through the hepatic artery. Moreover, the V-IO technique can also assist in diagnosis; liquid biopsy with selective venous sampling using a catheter-based approach has been reported in patients.71

Figure 2.

Figure 2

Examples of minimally invasive, image-guided interventional oncology procedures performed with catheter-directed and percutaneous approaches

(A) Arteriogram showing selective injection of Y90 microspheres in the anterior branch of the right hepatic artery. (B) Axial non-enhanced CT showing a microwave ablation of an apical left upper lobe nodule.

Percutaneous interventional oncology

Under image guidance, physicians can accurately place a needle into a tumor to perform diagnostic and therapeutic interventions. Percutaneous biopsy is an example of a diagnostic intervention that is used to collect specimens from the tumor for pathology and molecular interrogation.72 Ablation is an example of a therapeutic interventional procedure (Figure 2B) where percutaneous probes are used to deliver non-ionizing energy or other therapeutic agents to destroy the tumor in situ without surgical resection. Ablation is widely used for the treatment of malignancies in liver, kidney, bone, and lung.73, 74, 75, 76, 77, 78 Ablation techniques can be divided into two categories based on biophysical principles mediating the injury to the tumor, including chemical (ethanol or acetic acid) and non-ionizing energy-based ablation (thermal ablation and non-thermal ablation).79 This review will focus on the latter technique, which includes radiofrequency ablation (RFA), microwave ablation (MWA), cryotherapy (Cryo), and irreversible electroporation (IRE).

Locoregional CAR T therapy

Delivering immunotherapy directly into the tumor at the highest concentration may overcome problems with localization, infiltration, and systemic toxicity (Figure 3A). Over the past few decades, intraarterial chemotherapy has transformed the care of cancer patients using a minimally invasive image-guided approach. For example, intraarterial chemotherapy via the hepatic artery for liver colorectal metastasis has been shown to significantly improve the survival rate of patients compared with systemic therapy.80, 81, 82 This approach overcomes the disadvantages of intravenous systemic infusions, including systemic toxicity, first-pass metabolism, and non-targeted delivery. Interventional oncology procedures can therefore aid the efficacy of CAR T cell therapy by leveraging direct local delivery of the therapeutic (Figures 4A–4C). Locoregional delivery can have two specific advantages, namely (1) increasing delivery for greater local expansion and overcoming the issue of limited trafficking and (2) reducing the potential for on-target/off-tumor toxicity. Locoregional infusion of CAR T cells may also reduce on-target/off-tumor off-target toxicities. While severe colitis was observed via systemic infusion of CAR T cells targeting carcinoembryonic antigen (CEA), intraarterial infusion of TCR-modified T cells expressing anti-CEA TCR eliminated the incidence of grade 4 or 5 adverse events.83,84 Locoregional infusion potentially helps T cell trafficking. Direct infusion as well as transarterial infusion have been applied to metastatic liver tumors and metastatic breast cancer with evidence of intratumoral CAR T cells.84, 85, 86 Katz et al. showed an intratumoral presence of CAR T cells in patients 12 weeks after completing treatment for colorectal liver metastases.84 In addition, locoregional-infused CAR-T cells may expand locally and traffic safely to other tumor sites to promote further immune responses.87,88 Locoregional infusion presents several advantages, including increased hydrodynamic forces, improved T cell trafficking, and limited CAR T cell toxicity, by lowering the infused volume.87 Hydrodynamic force via transarterial infusion can overcome an elevated IFP that can inhibit CAR T cell migration through dense tumor stroma.26 Systemic infusion, however, theoretically cannot exceed intravascular pressure, which is typically lower than intratumoral IFP. Locoregional approaches for CAR T cell delivery have shown promising initial results.89, 90, 91, 92 Vitanza et al. recently reported a clinical study with locoregional infusion of HER2-specific CAR T cells for a central nervous system (CNS) tumor.93 They reported that administration of CAR T cells through a CNS catheter was feasible and tolerable. Therefore, randomized trials that compare systemic and locoregional infusion have to be performed to understand underlying mechanisms and identify comparative efficacy.

Figure 3.

Figure 3

Interventional oncology solutions for challenges in solid tumor immunotherapy

Figure 4.

Figure 4

Image-guided locoregional infusion of CAR T cells

(A) Arteriogram showing selective intraarterial infusion of CAR T cells through a 2.4 Fr microcatheter (black arrow) placed in the posterior branch of the right hepatic artery. (B) Axial non-enhanced CT showing intratumoral infusion of CAR T cells through a percutaneously placed 18 G needle in a right upper lobe pleural mesothelioma. (C) Direct infusion of CAR T cells in a 12 mm liver metastasis in segment VIII, shown here on non-enhanced axial CT.

Priming the TME for CAR T cell entry

Interventional oncology can deliver agents that can increase the infiltration or efficacy of systemically delivered CAR T cell in tumors. CAR T cell infiltration into solid tumors can be impeded from the lack of appropriate chemokines and other biological mediators. Therefore, priming the TME prior to infusion of CAR T cells can be another strategy to improve treatment outcomes (Figure 3B). Tumor ablation, regardless of the specific technique used, induces robust chemokine release and stimulates the localized inflammatory response and immune cell activation.94, 95, 96, 97, 98, 99, 100, 101, 102 Tumor ablation has also been shown to exhibit synergistic effects when combined with ICIs. Waitz et al. reported that combining Cryo with anti-CTLA-4 therapy induces anti-tumor immunity.94 Also, Zhao et al. reported that IRE has the potential to reverse resistance to immune checkpoint blockades.101 Likewise, TACE has been shown to promote immunogenic cell death and induce tumor-antigen-specific responses.103,104 As another V-IO treatment, SIRT may induce more immune cell infiltration when compared with TACE or surgery.105

The combination of these interventional oncology procedures, neo-adjuvant to CAR T cell, has not been investigated but presents several interesting synergies. Oncolytic virus (OV) is one of the other options for priming CAR T cell therapy. While systemic delivery of OV has demonstrated limited success,106 intratumoral delivery has advantages regarding lower systemic toxicity, less probability of inactivation by immune cells, and reduction of the administered viral load.107 Similar to ablation, the role of interventional oncology in OV delivery is not just injecting but also appropriate targeting with image modalities before and during the procedure.108 Another advantage of image-guided needle delivery is the possibility of monitoring targeted lesions and the ability to profile tumors by taking pretreatment samples. As for a larger tumor, a multipronged injection needle is available to reduce the number of injections in clinical studies.109, 110, 111 While injection procedures through needles have been the most investigated,112 several studies have provided the rationale for intraarterial regional delivery of OV.113, 114, 115 Combination of OV with CAR T cells showed promising results in preclinical studies,116, 117, 118 and one clinical trial is ongoing (ClinicalTrials.gov: NCT03740256). Similar to OV, interventional oncology can help prime TME for CAR T cells in many ways. Studies for combinational interventional oncology procedures with CAR T cells should be proposed.

Adjuvant strategies for CAR T cells via interventional oncology techniques

The efficacy of CAR T cells is often limited by immunosuppressive TMEs from the upregulation of PD-L1 and the presence of T regs, as well as other factors.24,119 This highlights the need for additional adjuvant strategies to support CAR T cells (Figure 3B). In fact, inadequate activation of tumor-specific CAR T cells has been reported in several clinical trials in solid tumors.42,120 Cytokine support is important for the proper activation of CAR T cells, as both interleukin (IL)-12 and IL-18 have been shown to enhance anti-tumor responses in preclinical models of solid tumors.121, 122, 123 However, the systemic infusion of cytokines has the potential to result in severe adverse events, as IL-12 systemic infusion resulted in severe toxicity.124 Historically, transarterial immunotherapy with interferon-gamma (IFNγ) and IL-2 followed by transarterial chemotherapy has been reported for HCC and unresectable colorectal liver metastasis in the 1990s.125,126 More recently, granulocyte macrophage colony-stimulating factor (GM-CSF) injection via a hepatic artery followed by chemoembolization was performed for patients with liver metastases of colorectal cancer and uveal melanoma.127, 128, 129 In these studies, GM-CSF was administered intraarterially to enhance local anti-tumor immune responses and reduce systemic side effects. Therefore, locoregional cytokine injections using interventional oncology techniques may be better tolerated in patients and may enhance anti-tumor responses during CAR T cell therapy.

Lymphodepletion was used prior to adoptive TIL transfer for the treatment of metastatic melanoma, leading to enhanced TIL homing and anti-tumor effects.130 The use of a regimen containing fludarabine and cyclophosphamide improved the overall response rate compared with patients without preconditioning, and this regimen has been subsequently adopted as part of the therapeutic strategy.131 Lymphodepletion prior to CAR T cell therapy also increased CAR T cell expansion and persistence, resulting in improved clinical outcomes.132, 133, 134 However, fludarabine is still potentially toxic for patients with renal failure or who are highly pretreated.135 Intraarterial chemotherapy, however, can increase the exposure time and the local concentrations of chemo-agents while reducing systemic toxicity.136, 137, 138, 139, 140 Therefore, locoregional chemotherapy theoretically provides locoregional lymphodepletion with less toxicity, resulting in good CAR T cell response with patients, especially those who are sensitive to fludarabine. Regional adjuvant cytokine delivery and lymphodepletion potentially activate CAR T cells and inactivate immunosuppressive cells. However, it is unclear whether it would recapitulate the homeostatic cytokine production or the innate immune activation due to damage to the gut epithelium. The comparison studies against systemic infusion also have not been performed yet. In addition, optimization of local drug delivery (dose, sustained release formulation, etc.) will be needed.

Monitoring T cell activity

CAR T cells are designed to recognize the target antigen, become activated, proliferate, and attack tumors after infusion into patients.141 Monitoring these steps is important for understanding the biological mechanisms that determine or influence in vivo efficacy.141,142 In some clinical trials, the sustained activity of the CAR T cell population is also considered an important predictor of anti-tumor efficacy.143,144 CAR T cell activity is currently monitored by detecting CAR T cells in peripheral venous blood sampled from upper limbs and in biopsy samples from local tumor sites.145 These clinical samples can be processed and analyzed with cytokine profile, flow cytometry, qPCR, or RNA sequencing (RNA-seq).22,30,145 These assays have potential limitations, as blood samples from systemic circulation or tumor cores may not properly represent the CAR T cell presence or activity in heterogeneous solid tumors. In addition to molecular biological assays, immunohistochemistry of tissue samples is commonly performed to understand the status of CAR T cells within the TME.86,146 The immunohistochemistry (IHC) method is commonly used to prove actual infiltration into solid tumors, but tumor tissue with heterogeneity does not provide accurate information about the temporal and spatial distribution of immune cells.147 Interventional oncology techniques could solve these limitations. Using V-IO, a catheter can be advanced close to the tumor site through the vessels. Selective venous sampling has the possibility of improving the diagnostic accuracy of liquid biopsy, as recently reported.71 Likewise, this method could be applied to collect samples to analyze CAR T cell activity with more CAR T cells and a more accurate condition than peripheral blood sampling. Using P-IO, physicians can perform multiple targeted biopsies. Conventional biopsies from the tumor core are affected by not only tumor heterogeneity but also necrotic avascular tumor tissue, which is unlikely to contain active immune cells. Biological hot spots in the tumor, however, can be peripheral, so multiple sampling at different sites should be appropriate.49 Multiple biopsies potentially increase the risk of complication, but interventional oncology physicians are experienced in evaluating anatomical accessibility, understanding the surrounding structure, and avoiding complications.148 Therefore, tumor biopsies can improve diagnostic accuracy with high information content and ease the negative effect of intertumoral heterogeneity. Together, V-IO and P-IO have the potential to provide more accurate and precise information in a minimally invasive manner.

Conclusions

Immunotherapy has grown to play a central role in oncology. Interventional oncology is an emerging modality for treating patients with cancer in a minimally invasive manner with unique features that can advance the utility and success of adoptive cell immunotherapy in solid tumors. This concept provides intriguing research opportunities with potential beneficial impacts on patients with what were historically considered to be incurable cancers. While preclinical data for combinational therapy using these two approaches appear promising, rigorous assessment of effectiveness with well-designed clinical trials must be performed to validate interventional oncology to advance CAR T cell immunotherapy of solid tumors.

Acknowledgments

P.S.A.’s laboratory work is supported by grants from the National Institutes of Health (P30 CA008748, R01 CA236615-01, and R01 CA235667); the U.S. Department of Defense (BC132124, LC160212, CA170630, and CA180889); the Baker Street Foundation; the Batishwa Fellowship; the Comedy vs Cancer Award; the Derfner Foundation; the Dalle Pezze Foundation; the Esophageal Cancer Education Fund; the Geoffrey Beene Foundation; the Memorial Sloan Kettering Technology Development Fund; the Miner Fund for Mesothelioma Research; Mr. William H. Goodwin and Alice Goodwin; the Commonwealth Foundation for Cancer Research; and the Experimental Therapeutics Center of Memorial Sloan Kettering Cancer Center. G.S. acknowledges grant and funding support from the National Cancer Institute of the National Institutes of Health under Award Number R01CA236615, the Dept. of Defense CDMRP PRCRP Award CA170630 and CA190888, and the Institute for Applied Life Sciences in the University of Massachusetts at Amherst. We thank Summer Koop of the Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, for editorial assistance.

Author contributions

Conceptualization, G.S., P.S.A., and S.B.S; investigation, Y.K. and M.G.; resources, Y.K., M.G., and W.C.; data curation, Y.K. and M.G.; writing – original draft preparation, G.S. and Y.K.; writing –review & editing, G.S., P.S.A., W.C., M.G., and S.B.S; supervision, G.S.; project administration, G.S.; funding acquisition, G.S., P.S.A., and S.B.S. All authors have read and agreed to the published version of the manuscript.

Declaration of interests

P.S.A. has received research funding from ATARA Biotherapeutics, has served on the Scientific Advisory Board or as consultant to ATARA Biotherapeutics, Bayer, Carisma Therapeutics, Imugene, ImmPACT Bio, and Takeda Therapeutics, and has patents, royalties, and intellectual property on mesothelin-targeted CARs and T cell therapies, which have been licensed to ATARA Biotherapeutics, as well as methods for the detection of cancer cells using virus, and pending patent applications on T cell therapies. S.B.S is a consultant to BTG, Johnson & Johnson, XACT, Adegro, and Medtronic. S.B.S. has funding support from GE Healthcare and Angiodynamics and holds stock in Aperture Medical. G.S. has received consulting fees from Farapulse and Intuitive Surgical and holds stock options in Aperture Medical. All other authors declare no competing interests. Memorial Sloan Kettering Cancer Center (MSK) has licensed intellectual property related to mesothelin-targeted CARs and T cell therapies to ATARA Biotherapeutics and has associated financial interests.

References

  • 1.Anderson K.G., Stromnes I.M., Greenberg P.D. Obstacles posed by the tumor microenvironment to T cell activity: a case for synergistic therapies. Cancer Cell. 2017;31:311–325. doi: 10.1016/j.ccell.2017.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Messerschmidt J.L., Prendergast G.C., Messerschmidt G.L. How cancers escape immune destruction and mechanisms of action for the new significantly active immune therapies: helping nonimmunologists decipher recent advances. The Oncologist. 2016;21:233–243. doi: 10.1634/theoncologist.2015-0282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Töpfer K., Kempe S., Müller N., Schmitz M., Bachmann M., Cartellieri M., Schackert G., Temme A. Tumor evasion from T cell surveillance. J. Biomed. Biotechnol. 2011;2011:e918471. doi: 10.1155/2011/918471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Farkona S., Diamandis E.P., Blasutig I.M. Cancer immunotherapy: the beginning of the end of cancer? BMC Med. 2016;14:73. doi: 10.1186/s12916-016-0623-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Zhang Y., Zhang Z. The history and advances in cancer immunotherapy: understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell. Mol. Immunol. 2020;17:807–821. doi: 10.1038/s41423-020-0488-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Haslam A., Prasad V. Estimation of the percentage of US patients with cancer who are eligible for and respond to checkpoint inhibitor immunotherapy drugs. JAMA Netw. Open. 2019;2:e192535. doi: 10.1001/jamanetworkopen.2019.2535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Yi M., Jiao D., Xu H., Liu Q., Zhao W., Han X., Wu K. Biomarkers for predicting efficacy of PD-1/PD-L1 inhibitors. Mol. Cancer. 2018;17:129. doi: 10.1186/s12943-018-0864-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Havel J.J., Chowell D., Chan T.A. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat. Rev. Cancer. 2019;19:133–150. doi: 10.1038/s41568-019-0116-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Fridman W.H., Pagès F., Sautès-Fridman C., Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat. Rev. Cancer. 2012;12:298–306. doi: 10.1038/nrc3245. [DOI] [PubMed] [Google Scholar]
  • 10.Tumeh P.C., Harview C.L., Yearley J.H., Shintaku I.P., Taylor E.J.M., Robert L., Chmielowski B., Spasic M., Henry G., Ciobanu V., et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515:568–571. doi: 10.1038/nature13954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Hegde P.S., Karanikas V., Evers S. The where, the when, and the how of immune monitoring for cancer immunotherapies in the era of checkpoint inhibition. Clin. Cancer Res. 2016;22:1865–1874. doi: 10.1158/1078-0432.CCR-15-1507. [DOI] [PubMed] [Google Scholar]
  • 12.Chen D.S., Mellman I. Elements of cancer immunity and the cancer–immune set point. Nature. 2017;541:321–330. doi: 10.1038/nature21349. [DOI] [PubMed] [Google Scholar]
  • 13.Bonaventura P., Shekarian T., Alcazer V., Valladeau-Guilemond J., Valsesia-Wittmann S., Amigorena S., Caux C., Depil S. Cold tumors: a therapeutic challenge for immunotherapy. Front. Immunol. 2019;10:168. doi: 10.3389/fimmu.2019.00168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Joseph R.W., Peddareddigari V.R., Liu P., Miller P.W., Overwijk W.W., Bekele N.B., Ross M.I., Lee J.E., Gershenwald J.E., Lucci A., et al. Impact of clinical and pathologic features on tumor-infiltrating lymphocyte expansion from surgically excised melanoma metastases for adoptive T-cell therapy. Clin. Cancer Res. 2011;17:4882–4891. doi: 10.1158/1078-0432.CCR-10-2769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Tran E., Turcotte S., Gros A., Robbins P.F., Lu Y.-C., Dudley M.E., Wunderlich J.R., Somerville R.P., Hogan K., Hinrichs C.S., et al. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science. 2014;344:641–645. doi: 10.1126/science.1251102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Tran E., Robbins P.F., Lu Y.-C., Prickett T.D., Gartner J.J., Jia L., Pasetto A., Zheng Z., Ray S., Groh E.M., et al. T-cell transfer therapy targeting mutant KRAS in cancer. N. Engl. J. Med. 2016;375:2255–2262. doi: 10.1056/NEJMoa1609279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Zacharakis N., Chinnasamy H., Black M., Xu H., Lu Y.-C., Zheng Z., Pasetto A., Langhan M., Shelton T., Prickett T., et al. Immune recognition of somatic mutations leading to complete durable regression in metastatic breast cancer. Nat. Med. 2018;24:724–730. doi: 10.1038/s41591-018-0040-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Govers C., Sebestyén Z., Coccoris M., Willemsen R.A., Debets R. T cell receptor gene therapy: strategies for optimizing transgenic TCR pairing. Trends Mol. Med. 2010;16:77–87. doi: 10.1016/j.molmed.2009.12.004. [DOI] [PubMed] [Google Scholar]
  • 19.Essand M., Loskog A.S.I. Genetically engineered T cells for the treatment of cancer. J. Intern. Med. 2013;273:166–181. doi: 10.1111/joim.12020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Li D., Li X., Zhou W.-L., Huang Y., Liang X., Jiang L., Yang X., Sun J., Li Z., Han W.-D., et al. Genetically engineered T cells for cancer immunotherapy. Signal Transduct. Target Ther. 2019;4:35. doi: 10.1038/s41392-019-0070-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Ramos C.A., Dotti G. Chimeric antigen receptor (CAR)-Engineered lymphocytes for cancer therapy. Expert Opin. Biol. Ther. 2011;11:855–873. doi: 10.1517/14712598.2011.573476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Maude S.L., Frey N., Shaw P.A., Aplenc R., Barrett D.M., Bunin N.J., Chew A., Gonzalez V.E., Zheng Z., Lacey S.F., et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 2014;371:1507–1517. doi: 10.1056/NEJMoa1407222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Neelapu S.S., Locke F.L., Bartlett N.L., Lekakis L.J., Miklos D.B., Jacobson C.A., Braunschweig I., Oluwole O.O., Siddiqi T., Lin Y., et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N. Engl. J. Med. 2017;377:2531–2544. doi: 10.1056/NEJMoa1707447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Lanitis E., Dangaj D., Irving M., Coukos G. Mechanisms regulating T-cell infiltration and activity in solid tumors. Ann. Oncol. 2017;28:xii18–xii32. doi: 10.1093/annonc/mdx238. [DOI] [PubMed] [Google Scholar]
  • 25.Xia A.-L., Wang X.-C., Lu Y.-J., Lu X.-J., Sun B. Chimeric-antigen receptor T (CAR-T) cell therapy for solid tumors: challenges and opportunities. Oncotarget. 2017;8:90521–90531. doi: 10.18632/oncotarget.19361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Hardaway J.C., Prince E., Arepally A., Katz S.C. Regional infusion of chimeric antigen receptor T cells to overcome barriers for solid tumor immunotherapy. J. Vasc. Interv. Radiol. 2018;29:1017–1021.e1. doi: 10.1016/j.jvir.2018.03.001. [DOI] [PubMed] [Google Scholar]
  • 27.Haanen J.B.a.G., Carbonnel F., Robert C., Kerr K.M., Peters S., Larkin J., Jordan K. Management of toxicities from immunotherapy: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2017;28:iv119–iv142. doi: 10.1093/annonc/mdx225. [DOI] [PubMed] [Google Scholar]
  • 28.Brudno J.N., Kochenderfer J.N. Recent advances in CAR T-cell toxicity: mechanisms, manifestations and management. Blood Rev. 2019;34:45–55. doi: 10.1016/j.blre.2018.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Scarfò I., Maus M.V. Current approaches to increase CAR T cell potency in solid tumors: targeting the tumor microenvironment. J. Immunother. Cancer. 2017;5:28. doi: 10.1186/s40425-017-0230-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Hu Y., Huang J. The chimeric antigen receptor detection toolkit. Front. Immunol. 2020;11:1770. doi: 10.3389/fimmu.2020.01770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Martínez Bedoya D., Dutoit V., Migliorini D. Allogeneic CAR T cells: an alternative to overcome challenges of CAR T cell therapy in glioblastoma. Front. Immunol. 2021;12:640082. doi: 10.3389/fimmu.2021.640082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Garg A.D., Krysko D.V., Verfaillie T., Kaczmarek A., Ferreira G.B., Marysael T., Rubio N., Firczuk M., Mathieu C., Roebroek A.J.M., et al. A novel pathway combining calreticulin exposure and ATP secretion in immunogenic cancer cell death. EMBO J. 2012;31:1062–1079. doi: 10.1038/emboj.2011.497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Senovilla L., Vitale I., Martins I., Tailler M., Pailleret C., Michaud M., Galluzzi L., Adjemian S., Kepp O., Niso-Santano M., et al. An immunosurveillance mechanism controls cancer cell ploidy. Science. 2012;337:1678–1684. doi: 10.1126/science.1224922. [DOI] [PubMed] [Google Scholar]
  • 34.Ma Y., Adjemian S., Mattarollo S.R., Yamazaki T., Aymeric L., Yang H., Portela Catani J.P., Hannani D., Duret H., Steegh K., et al. Anticancer chemotherapy-induced intratumoral recruitment and differentiation of antigen-presenting cells. Immunity. 2013;38:729–741. doi: 10.1016/j.immuni.2013.03.003. [DOI] [PubMed] [Google Scholar]
  • 35.Muranski P., Boni A., Wrzesinski C., Citrin D.E., Rosenberg S.A., Childs R., Restifo N.P. Increased intensity lymphodepletion and adoptive immunotherapy—how far can we go? Nat. Clin. Pract. Oncol. 2006;3:668–681. doi: 10.1038/ncponc0666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Reits E.A., Hodge J.W., Herberts C.A., Groothuis T.A., Chakraborty M., Wansley E., Camphausen K., Luiten R.M., de Ru A.H., Neijssen J., et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J. Exp. Med. 2006;203:1259–1271. doi: 10.1084/jem.20052494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Matsumura S., Wang B., Kawashima N., Braunstein S., Badura M., Cameron T.O., Babb J.S., Schneider R.J., Formenti S.C., Dustin M.L., et al. Radiation-induced CXCL16 release by breast cancer cells attracts effector T cells. J. Immunol. 2008;181:3099–3107. doi: 10.4049/jimmunol.181.5.3099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Liao Y.-P., Wang C.-C., Butterfield L.H., Economou J.S., Ribas A., Meng W.S., Iwamoto K.S., McBride W.H. Ionizing radiation affects human MART-1 melanoma antigen processing and presentation by dendritic cells. J. Immunol. 2004;173:2462–2469. doi: 10.4049/jimmunol.173.4.2462. [DOI] [PubMed] [Google Scholar]
  • 39.Radford I.R. Radiation response of mouse lymphoid and myeloid cell lines. Part I. Sensitivity to killing by ionizing radiation, rate of loss of viability, and cell type of origin. Int. J. Radiat. Biol. 1994;65:203–215. doi: 10.1080/09553009414550241. [DOI] [PubMed] [Google Scholar]
  • 40.Palayoor S.T., Macklis R.M., Bump E.A., Coleman C.N. Modulation of radiation-induced apoptosis and G2/M block in murine T-lymphoma cells. Radiat. Res. 1995;141:235–243. [PubMed] [Google Scholar]
  • 41.John L.B., Devaud C., Duong C.P.M., Yong C.S., Beavis P.A., Haynes N.M., Chow M.T., Smyth M.J., Kershaw M.H., Darcy P.K. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin. Cancer Res. 2013;19:5636–5646. doi: 10.1158/1078-0432.CCR-13-0458. [DOI] [PubMed] [Google Scholar]
  • 42.Gargett T., Yu W., Dotti G., Yvon E.S., Christo S.N., Hayball J.D., Lewis I.D., Brenner M.K., Brown M.P. GD2-specific CAR T cells undergo potent activation and deletion following antigen encounter but can be protected from activation-induced cell death by PD-1 blockade. Mol. Ther. 2016;24:1135–1149. doi: 10.1038/mt.2016.63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Cherkassky, L., Morello, A., Villena-Vargas, J., Feng, Y., Dimitrov, D.S., Jones, D.R., Sadelain, M., and Adusumilli, P.S. Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition. J. Clin. Invest. 126, 3130–3144. [DOI] [PMC free article] [PubMed]
  • 44.Burga R.A., Thorn M., Point G.R., Guha P., Nguyen C.T., Licata L.A., DeMatteo R.P., Ayala A., Espat N.J., Junghans R.P., et al. Liver myeloid-derived suppressor cells expand in response to liver metastases in mice and inhibit the anti-tumor efficacy of anti-CEA CAR-T. Cancer Immunol. Immunother. 2015;64:817–829. doi: 10.1007/s00262-015-1692-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Chong E.A., Melenhorst J.J., Lacey S.F., Ambrose D.E., Gonzalez V., Levine B.L., June C.H., Schuster S.J. PD-1 blockade modulates chimeric antigen receptor (CAR)–modified T cells: refueling the CAR. Blood. 2017;129:1039–1041. doi: 10.1182/blood-2016-09-738245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Heczey A., Louis C.U., Savoldo B., Dakhova O., Durett A., Grilley B., Liu H., Wu M.F., Mei Z., Gee A., et al. CAR T cells administered in combination with lymphodepletion and PD-1 inhibition to patients with neuroblastoma. Mol. Ther. 2017;25:2214–2224. doi: 10.1016/j.ymthe.2017.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Matsui O., Miyayama S., Sanada J., Kobayashi S., Khoda W., Minami T., Kozaka K., Gabata T. Interventional oncology: new options for interstitial treatments and intravascular approaches. J. Hepato-Biliary-Pancreatic Sci. 2010;17:407–409. doi: 10.1007/s00534-009-0234-z. [DOI] [PubMed] [Google Scholar]
  • 48.Kim H.S., Chapiro J., Geschwind J.-F.H. From the guest editor: interventional oncology: the fourth pillar of oncology. Cancer J. 2016;22:363–364. doi: 10.1097/PPO.0000000000000235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Schoenberg S.O., Attenberger U.I., Solomon S.B., Weissleder R. Developing a roadmap for interventional oncology. Oncologist. 2018;23:1162–1170. doi: 10.1634/theoncologist.2017-0654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Raoul J.-L., Forner A., Bolondi L., Cheung T.T., Kloeckner R., Baere T. Updated use of TACE for hepatocellular carcinoma treatment: how and when to use it based on clinical evidence. Cancer Treat. Rev. 2019;72:28–36. doi: 10.1016/j.ctrv.2018.11.002. [DOI] [PubMed] [Google Scholar]
  • 51.European Association for the Study of the Liver EASL clinical practice guidelines: management of hepatocellular carcinoma. J. Hepatol. 2018;69:182–236. doi: 10.1016/j.jhep.2018.03.019. [DOI] [PubMed] [Google Scholar]
  • 52.Padia S.A. Y90 clinical data update: cholangiocarcinoma, neuroendocrine tumor, melanoma, and breast cancer metastatic disease. Tech. Vasc. Interv. Radiol. 2019;22:81–86. doi: 10.1053/j.tvir.2019.02.008. [DOI] [PubMed] [Google Scholar]
  • 53.Hu J., Albadawi H., Oklu R., Chong B.W., Deipolyi A.R., Sheth R.A., Khademhosseini A. Advances in biomaterials and technologies for vascular embolization. Adv. Mater. 2019;31:e1901071. doi: 10.1002/adma.201901071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Romaric L., Boris G., Jean-Pierre C., Denis K. Endovascular therapeutic embolisation: an overview of occluding agents and their effects on embolised tissues. Curr. Vasc. Pharmacol. 2009;7:250–263. doi: 10.2174/157016109787455617. [DOI] [PubMed] [Google Scholar]
  • 55.Kishore S.A., Bajwa R., Madoff D.C. Embolotherapeutic strategies for hepatocellular carcinoma: 2020 update. Cancers. 2020;12:791. doi: 10.3390/cancers12040791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Ghavimi S., Apfel T., Azimi H., Persaud A., Pyrsopoulos N.T. Management and treatment of hepatocellular carcinoma with immunotherapy: a review of current and future options. J. Clin. Transl Hepatol. 2020;8:168–176. doi: 10.14218/JCTH.2020.00001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Chang Y., Jeong S.W., Young Jang J., Jae Kim Y. Recent updates of transarterial chemoembolilzation in hepatocellular carcinoma. Int. J. Mol. Sci. 2020;21:8165. doi: 10.3390/ijms21218165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Gunn A.J., Patel A.R., Rais-Bahrami S. Role of angio-embolization for renal cell carcinoma. Curr. Urol. Rep. 2018;19:76. doi: 10.1007/s11934-018-0827-7. [DOI] [PubMed] [Google Scholar]
  • 59.Ginat D.T., Saad W.E.A., Turba U.C. Transcatheter renal artery embolization for management of renal and adrenal tumors. Tech. Vasc. Interv. Radiol. 2010;13:75–88. doi: 10.1053/j.tvir.2010.02.003. [DOI] [PubMed] [Google Scholar]
  • 60.Cordovilla R., Bollo de Miguel E., Nuñez Ares A., Cosano Povedano F.J., Herráez Ortega I., Jiménez Merchán R. Diagnosis and treatment of hemoptysis. Arch. Bronconeumol. 2016;52:368–377. doi: 10.1016/j.arbres.2015.12.002. [DOI] [PubMed] [Google Scholar]
  • 61.Cho S.B., Hur S., Kim H.-C., Jae H.J., Lee M., Kim M., Kim J.-E., Lee J.H., Chung J.W. Transcatheter arterial embolization for advanced gastric cancer bleeding. Medicine. 2020;99:e19630. doi: 10.1097/MD.0000000000019630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Lai E.C.H., Lau W.Y. Spontaneous rupture of hepatocellular carcinoma: a systematic review. Arch. Surg. 2006;141:191–198. doi: 10.1001/archsurg.141.2.191. [DOI] [PubMed] [Google Scholar]
  • 63.Omid-Fard N., Fisher C.G., Heran M.K. The evolution of pre-operative spine tumour embolization. Br. J. Radiol. 2019;92:20180899. doi: 10.1259/bjr.20180899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Ellis J.A., D’Amico R., Sisti M.B., Bruce J.N., McKhann G.M., Lavine S.D., Meyers P.M., Strozyk D. Pre-operative intracranial meningioma embolization. Expert Rev. Neurother. 2011;11:545–556. doi: 10.1586/ern.11.29. [DOI] [PubMed] [Google Scholar]
  • 65.Ma J., Tullius T., Van Ha T.G. Update on preoperative embolization of bone metastases. Semin. Intervent. Radiol. 2019;36:241–248. doi: 10.1055/s-0039-1693120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Shah A., Choudhri O., Jung H., Li G. Preoperative endovascular embolization of meningiomas: update on therapeutic options. Neurosurg. Focus. 2015;38:E7. doi: 10.3171/2014.12.FOCUS14728. [DOI] [PubMed] [Google Scholar]
  • 67.Vogl T.J., Gruber-Rouh T. HCC: transarterial therapies—what the interventional radiologist can offer. Dig. Dis. Sci. 2019;64:959–967. doi: 10.1007/s10620-019-05542-5. [DOI] [PubMed] [Google Scholar]
  • 68.Van Thai N., Thinh N.T., Ky T.D., Bang M.H., Giang D.T., Ha L.N., Son M.H., Tien D.D., Lee H.W. Efficacy and safety of selective internal radiation therapy with yttrium-90 for the treatment of unresectable hepatocellular carcinoma. BMC Gastroenterol. 2021;21:216. doi: 10.1186/s12876-021-01805-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Townsend A.R., Chong L.C., Karapetis C., Price T.J. Selective internal radiation therapy for liver metastases from colorectal cancer. Cancer Treat. Rev. 2016;50:148–154. doi: 10.1016/j.ctrv.2016.09.007. [DOI] [PubMed] [Google Scholar]
  • 70.Barbier C.E., Garske-Román U., Sandström M., Nyman R., Granberg D. Selective internal radiation therapy in patients with progressive neuroendocrine liver metastases. Eur. J. Nucl. Med. Mol. Imaging. 2016;43:1425–1431. doi: 10.1007/s00259-015-3264-6. [DOI] [PubMed] [Google Scholar]
  • 71.Damascelli B., Tichà V., Repetti E., Dorji T. Beyond standard practice in liquid biopsy: selective venous sampling. J. Vasc. Interv. Radiol. 2021;32:668–671. doi: 10.1016/j.jvir.2021.02.010. [DOI] [PubMed] [Google Scholar]
  • 72.Pritzker K.P.H., Nieminen H.J. Needle biopsy adequacy in the era of precision medicine and value-based Health care. Arch. Pathol. Lab. Med. 2019;143:1399–1415. doi: 10.5858/arpa.2018-0463-RA. [DOI] [PubMed] [Google Scholar]
  • 73.Ahmed M., Solbiati L., Brace C.L., Breen D.J., Callstrom M.R., Charboneau J.W., Chen M.-H., Choi B.I., de Baère T., Dodd G.D., et al. Image-guided tumor ablation: standardization of terminology and reporting criteria—a 10-year update. J. Vasc. Interv. Radiol. 2014;25:1691–1705.e4. doi: 10.1016/j.jvir.2014.08.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.N’Kontchou G., Mahamoudi A., Aout M., Ganne-Carrié N., Grando V., Coderc E., Vicaut E., Trinchet J.C., Sellier N., Beaugrand M., et al. Radiofrequency ablation of hepatocellular carcinoma: long-term results and prognostic factors in 235 Western patients with cirrhosis. Hepatology. 2009;50:1475–1483. doi: 10.1002/hep.23181. [DOI] [PubMed] [Google Scholar]
  • 75.Prud’homme C., Deschamps F., Moulin B., Hakime A., Al-Ahmar M., Moalla S., Roux C., Teriitehau C., Baere T., Tselikas L. Image-guided lung metastasis ablation: a literature review. Int. J. Hyperthermia. 2019;36:37–45. doi: 10.1080/02656736.2019.1647358. [DOI] [PubMed] [Google Scholar]
  • 76.Venturini M., Cariati M., Marra P., Masala S., Pereira P.L., Carrafiello G. CIRSE standards of practice on thermal ablation of primary and secondary lung tumours. Cardiovasc. Intervent. Radiol. 2020;43:667–683. doi: 10.1007/s00270-020-02432-6. [DOI] [PubMed] [Google Scholar]
  • 77.Filippiadis D., Mauri G., Marra P., Charalampopoulos G., Gennaro N., Cobelli F.D. Percutaneous ablation techniques for renal cell carcinoma: current status and future trends. Int. J. Hyperthermia. 2019;36:21–30. doi: 10.1080/02656736.2019.1647352. [DOI] [PubMed] [Google Scholar]
  • 78.Moynagh M.R., Kurup A.N., Callstrom M.R. Thermal ablation of bone metastases. Semin. Intervent. Radiol. 2018;35:299–308. doi: 10.1055/s-0038-1673422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Ahmed M., Solbiati L., Brace C.L., Breen D.J., Callstrom M.R., Charboneau J.W., Chen M.-H., Choi B.I., de Baère T., Dodd G.D., et al. Image-guided tumor ablation: standardization of terminology and reporting criteria—a 10-year update. Radiology. 2014;273:241–260. doi: 10.1148/radiol.14132958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kemeny N.E., Niedzwiecki D., Hollis D.R., Lenz H.-J., Warren R.S., Naughton M.J., Weeks J.C., Sigurdson E.R., Herndon J.E., Zhang C., et al. Hepatic arterial infusion versus systemic therapy for hepatic metastases from colorectal cancer: a randomized trial of efficacy, quality of life, and molecular markers (CALGB 9481) JCO. 2006;24:1395–1403. doi: 10.1200/JCO.2005.03.8166. [DOI] [PubMed] [Google Scholar]
  • 81.Lewandowski R.J., Geschwind J.-F., Liapi E., Salem R. Transcatheter intraarterial therapies: rationale and overview. Radiology. 2011;259:641–657. doi: 10.1148/radiol.11081489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.D’Angelica M.I., Correa-Gallego C., Paty P.B., Cercek A., Gewirtz A.N., Chou J.F., Capanu M., Kingham T.P., Fong Y., DeMatteo R.P., et al. Phase II trial of hepatic artery infusional and systemic chemotherapy for patients with unresectable hepatic metastases from colorectal cancer: conversion to resection and long-term outcomes. Ann. Surg. 2015;261:353–360. doi: 10.1097/SLA.0000000000000614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Parkhurst M.R., Yang J.C., Langan R.C., Dudley M.E., Nathan D.-A.N., Feldman S.A., Davis J.L., Morgan R.A., Merino M.J., Sherry R.M., et al. T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol. Ther. 2011;19:620–626. doi: 10.1038/mt.2010.272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Katz S.C., Burga R.A., McCormack E., Wang L.J., Mooring W., Point G.R., Khare P.D., Thorn M., Ma Q., Stainken B.F., 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 Res. 2015;21:3149–3159. doi: 10.1158/1078-0432.CCR-14-1421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Priceman S.J., Tilakawardane D., Jeang B., Aguilar B., Murad J.P., Park A.K., Chang W.-C., Ostberg J.R., Neman J., Jandial R., et al. Regional delivery of chimeric antigen receptor–engineered T cells effectively targets HER2+ breast cancer metastasis to the brain. Clin. Cancer Res. 2018;24:95–105. doi: 10.1158/1078-0432.CCR-17-2041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Tchou J., Zhao Y., Levine B.L., Zhang P.J., Davis M.M., Melenhorst J.J., Kulikovskaya I., Brennan A.L., Liu X., Lacey S.F., et al. Safety and efficacy of intratumoral injections of chimeric antigen receptor (CAR) T cells in metastatic breast cancer. Cancer Immunol. Res. 2017;5:1152–1161. doi: 10.1158/2326-6066.CIR-17-0189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Beatty G.L., Haas A.R., Maus M.V., Torigian D.A., Soulen M.C., Plesa G., Chew A., Zhao Y., Levine B.L., Albelda S.M., et al. Mesothelin-specific chimeric antigen receptor mRNA-engineered T cells induce anti-tumor activity in solid malignancies. Cancer Immunol. Res. 2014;2:112–120. doi: 10.1158/2326-6066.CIR-13-0170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Adusumilli P.S., Cherkassky L., Villena-Vargas J., Colovos C., Servais E., Plotkin J., Jones D.R., Sadelain M. Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunity. Sci. Transl. Med. 2014;6:261ra151. doi: 10.1126/scitranslmed.3010162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Adusumilli P.S., Zauderer M.G., Riviere I., Solomon S.B., Rusch V.W., O’Cearbhaill R.E., Zhu A., Cheema W., Chintala N.K., Halton E., 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;11:2748–2763. doi: 10.1158/2159-8290.CD-21-0407. published online ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Sridhar P., Petrocca F. Regional delivery of chimeric antigen receptor (CAR) T-cells for cancer therapy. Cancers. 2017;9:92. doi: 10.3390/cancers9070092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Theruvath J., Sotillo E., Mount C.W., Graef C.M., Delaidelli A., Heitzeneder S., Labanieh L., Dhingra S., Leruste A., Majzner R.G., et al. Locoregionally administered B7-H3-targeted CAR T cells for treatment of atypical teratoid/rhabdoid tumors. Nat. Med. 2020;26:712–719. doi: 10.1038/s41591-020-0821-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Donovan L.K., Delaidelli A., Joseph S.K., Bielamowicz K., Fousek K., Holgado B.L., Manno A., Srikanthan D., Gad A.Z., Van Ommeren R., et al. Locoregional delivery of CAR T cells to the cerebrospinal fluid for treatment of metastatic medulloblastoma and ependymoma. Nat. Med. 2020;26:720–731. doi: 10.1038/s41591-020-0827-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Vitanza N.A., Johnson A.J., Wilson A.L., Brown C., Yokoyama J.K., Künkele A., Chang C.A., Rawlings-Rhea S., Huang W., Seidel K., 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;27:1544–1552. doi: 10.1038/s41591-021-01404-8. [DOI] [PubMed] [Google Scholar]
  • 94.Waitz R., Solomon S.B., Petre E.N., Trumble A.E., Fassò M., Norton L., Allison J.P. Potent induction of tumor immunity by combining tumor cryoablation with anti-CTLA-4 therapy. Cancer Res. 2012;72:430–439. doi: 10.1158/0008-5472.CAN-11-1782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Lu P., Zhu X.-Q., Xu Z.-L., Zhou Q., Zhang J., Wu F. Increased infiltration of activated tumor-infiltrating lymphocytes after high intensity focused ultrasound ablation of human breast cancer. Surgery. 2009;145:286–293. doi: 10.1016/j.surg.2008.10.010. [DOI] [PubMed] [Google Scholar]
  • 96.Wissniowski T.T., Hänsler J., Neureiter D., Frieser M., Schaber S., Esslinger B., Voll R., Strobel D., Hahn E.G., Schuppan D. Activation of tumor-specific T lymphocytes by radio-frequency ablation of the VX2 hepatoma in rabbits. Cancer Res. 2003;63:6496–6500. [PubMed] [Google Scholar]
  • 97.Matin S.F., Sharma P., Gill I.S., Tannenbaum C., Hobart M.G., Novick A.C., Finke J.H. Immunological response to renal cryoablation in an in vivo orthotopic renal cell carcinoma murine model. J. Urol. 2009;183:333–338. doi: 10.1016/j.juro.2009.08.110. [DOI] [PubMed] [Google Scholar]
  • 98.White S.B., Zhang Z., Chen J., Gogineni V.R., Larson A.C. Early immunologic response of irreversible electroporation versus cryoablation in a rodent model of pancreatic cancer. J. Vasc. Interv. Radiol. 2018;29:1764–1769. doi: 10.1016/j.jvir.2018.07.009. [DOI] [PubMed] [Google Scholar]
  • 99.Chu K.F., Dupuy D.E. Thermal ablation of tumours: biological mechanisms and advances in therapy. Nat. Rev. Cancer. 2014;14:199–208. doi: 10.1038/nrc3672. [DOI] [PubMed] [Google Scholar]
  • 100.Li X., Xu K., Li W., Qiu X., Ma B., Fan Q., Li Z. Immunologic response to tumor ablation with irreversible electroporation. PLoS One. 2012;7:e48749. doi: 10.1371/journal.pone.0048749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Zhao J., Wen X., Tian L., Li T., Xu C., Wen X., Melancon M.P., Gupta S., Shen B., Peng W., et al. Irreversible electroporation reverses resistance to immune checkpoint blockade in pancreatic cancer. Nat. Commun. 2019;10:899. doi: 10.1038/s41467-019-08782-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Zerbini A., Pilli M., Penna A., Pelosi G., Schianchi C., Molinari A., Schivazappa S., Zibera C., Fagnoni F.F., Ferrari C., et al. Radiofrequency thermal ablation of hepatocellular carcinoma liver nodules can activate and enhance tumor-specific T-cell responses. Cancer Res. 2006;66:1139–1146. doi: 10.1158/0008-5472.CAN-05-2244. [DOI] [PubMed] [Google Scholar]
  • 103.Kohles N., Nagel D., Jüngst D., Stieber P., Holdenrieder S. Predictive value of immunogenic cell death biomarkers HMGB1, sRAGE, and DNase in liver cancer patients receiving transarterial chemoembolization therapy. Tumor Biol. 2012;33:2401–2409. doi: 10.1007/s13277-012-0504-2. [DOI] [PubMed] [Google Scholar]
  • 104.Ayaru L., Pereira S.P., Alisa A., Pathan A.A., Williams R., Davidson B., Burroughs A.K., Meyer T., Behboudi S. Unmasking of α-fetoprotein-specific CD4+ T cell responses in hepatocellular carcinoma patients undergoing embolization. J. Immunol. 2007;178:1914–1922. doi: 10.4049/jimmunol.178.3.1914. [DOI] [PubMed] [Google Scholar]
  • 105.Craciun L., de Wind R., Demetter P., Lucidi V., Bohlok A., Michiels S., Bouazza F., Vouche M., Tancredi I., Verset G., et al. Retrospective analysis of the immunogenic effects of intra-arterial locoregional therapies in hepatocellular carcinoma: a rationale for combining selective internal radiation therapy (SIRT) and immunotherapy. BMC Cancer. 2020;20:135. doi: 10.1186/s12885-020-6613-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Seymour L.W., Fisher K.D. Oncolytic viruses: finally delivering. Br. J. Cancer. 2016;114:357–361. doi: 10.1038/bjc.2015.481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Yokoda R., Nagalo B.M., Vernon B., Oklu R., Albadawi H., DeLeon T.T., Zhou Y., Egan J.B., Duda D.G., Borad M.J. Oncolytic virus delivery: from nano-pharmacodynamics to enhanced oncolytic effect. Oncolytic Virother. 2017;6:39–49. doi: 10.2147/OV.S145262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Goldberg S.N., Grassi C.J., Cardella J.F., Charboneau J.W., Dodd G.D., Dupuy D.E., Gervais D.A., Gillams A.R., Kane R.A., Lee F.T., et al. Image-guided tumor ablation: standardization of terminology and reporting criteria. Radiology. 2005;235:728–739. doi: 10.1148/radiol.2353042205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Zeh H.J., Downs-Canner S., McCart J.A., Guo Z.S., Rao U.N.M., Ramalingam L., Thorne S.H., Jones H.L., Kalinski P., Wieckowski E., et al. First-in-man study of western reserve strain oncolytic vaccinia virus: safety, systemic spread, and antitumor activity. Mol. Ther. 2015;23:202–214. doi: 10.1038/mt.2014.194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Heo J., Reid T., Ruo L., Breitbach C.J., Rose S., Bloomston M., Cho M., Lim H.Y., Chung H.C., Kim C.W., et al. Randomized dose-finding clinical trial of oncolytic immunotherapeutic vaccinia JX-594 in liver cancer. Nat. Med. 2013;19:329–336. doi: 10.1038/nm.3089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Streby K.A., Geller J.I., Currier M.A., Warren P.S., Racadio J.M., Towbin A.J., Vaughan M.R., Triplet M., Ott-Napier K., Dishman D.J., et al. Intratumoral injection of HSV1716, an oncolytic herpes virus, is safe and shows evidence of immune response and viral replication in young cancer patients. Clin. Cancer Res. 2017;23:3566–3574. doi: 10.1158/1078-0432.CCR-16-2900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Li L., Liu S., Han D., Tang B., Ma J. Delivery and biosafety of oncolytic virotherapy. Front. Oncol. 2020;10:475. doi: 10.3389/fonc.2020.00475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Reid T., Galanis E., Abbruzzese J., Sze D., Wein L.M., Andrews J., Randlev B., Heise C., Uprichard M., Hatfield M., et al. Hepatic arterial infusion of a replication-selective oncolytic adenovirus (dl1520): phase II viral, immunologic, and clinical endpoints. Cancer Res. 2002;62:6070–6079. [PubMed] [Google Scholar]
  • 114.Li Y., Li L.-J., Wang L.-J., Zhang Z., Gao N., Liang C.-Y., Huang Y.-D., Han B. Selective intra-arterial infusion of rAd-p53 with chemotherapy for advanced oral cancer: a randomized clinical trial. BMC Med. 2014;12:16. doi: 10.1186/1741-7015-12-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Srinivasan V.M., Lang F.F., Kan P. Intraarterial delivery of virotherapy for glioblastoma. Neurosurg. Focus. 2021;50:E7. doi: 10.3171/2020.11.FOCUS20845. [DOI] [PubMed] [Google Scholar]
  • 116.Watanabe, K., Luo, Y., Da, T., Guedan, S., Ruella, M., Scholler, J., Keith, B., Young, R.M., Engels, B., Sorsa, S., et al. Pancreatic cancer therapy with combined mesothelin-redirected chimeric antigen receptor T cells and cytokine-armed oncolytic adenoviruses. JCI Insight 3, e99573. [DOI] [PMC free article] [PubMed]
  • 117.Tanoue K., Shaw A.R., Watanabe N., Porter C., Rana B., Gottschalk S., Brenner M., Suzuki M. Armed oncolytic adenovirus expressing PD-L1 mini-body enhances anti-tumor effects of chimeric antigen receptor T-cells in solid tumors. Cancer Res. 2017;77:2040–2051. doi: 10.1158/0008-5472.CAN-16-1577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Rosewell Shaw A., Porter C.E., Watanabe N., Tanoue K., Sikora A., Gottschalk S., Brenner M.K., Suzuki M. Adenovirotherapy delivering cytokine and checkpoint inhibitor augments CAR T cells against metastatic head and neck cancer. Mol. Ther. 2017;25:2440–2451. doi: 10.1016/j.ymthe.2017.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Schreiber R.D., Old L.J., Smyth M.J. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science. 2011;331:1565–1570. doi: 10.1126/science.1203486. [DOI] [PubMed] [Google Scholar]
  • 120.O’Rourke D.M., Nasrallah M.P., Desai A., Melenhorst J.J., Mansfield K., Morrissette J.J.D., Martinez-Lage M., Brem S., Maloney E., Shen A., et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci. Transl. Med. 2017;9:eaaa0984. doi: 10.1126/scitranslmed.aaa0984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Koneru M., Purdon T.J., Spriggs D., Koneru S., Brentjens R.J. IL-12 secreting tumor-targeted chimeric antigen receptor T cells eradicate ovarian tumors in vivo. Oncoimmunology. 2015;4:e994446. doi: 10.4161/2162402X.2014.994446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Chmielewski M., Abken H. CAR T cells releasing IL-18 convert to T-Bethigh FoxO1low effectors that exhibit augmented activity against advanced solid tumors. Cell Rep. 2017;21:3205–3219. doi: 10.1016/j.celrep.2017.11.063. [DOI] [PubMed] [Google Scholar]
  • 123.Hu B., Ren J., Luo Y., Keith B., Young R.M., Scholler J., Zhao Y., June C.H. Augmentation of antitumor immunity by human and mouse CAR T cells secreting IL-18. Cell Rep. 2017;20:3025–3033. doi: 10.1016/j.celrep.2017.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Leonard J.P., Sherman M.L., Fisher G.L., Buchanan L.J., Larsen G., Atkins M.B., Sosman J.A., Dutcher J.P., Vogelzang N.J., Ryan J.L. Effects of single-dose interleukin-12 exposure on interleukin-12–associated toxicity and interferon-γ production. Blood. 1997;90:2541–2548. [PubMed] [Google Scholar]
  • 125.Lygidakis N.J., Kosmidis P., Ziras N., Parissis J., Kyparidou E. Combined transarterial targeting locoregional immunotherapy-chemotherapy for patients with unresectable hepatocellular carcinoma: a new alternative for an old problem. J. Interferon Cytokine Res. 1995;15:467–472. doi: 10.1089/jir.1995.15.467. [DOI] [PubMed] [Google Scholar]
  • 126.Lygidakis N.J., Savanis G., Pothoulakis J., Kapetanakis A. Transarterial locoregional immunostimulation and chemotherapy in patients with unresectable secondary liver tumours. Anticancer Res. 1994;14:643–646. [PubMed] [Google Scholar]
  • 127.Müller H., Nakchbandi W., Chatzissavvidis I., Valek V. Intra-arterial infusion of 5-fluorouracil plus granulocyte–macrophage colony-stimulating factor (GM-CSF) and chemoembolization with melphalan in the treatment of disseminated colorectal liver metastases. Eur. J. Surg. Oncol. 2001;27:652–661. doi: 10.1053/ejso.2001.1193. [DOI] [PubMed] [Google Scholar]
  • 128.Sato T., Eschelman D.J., Gonsalves C.F., Terai M., Chervoneva I., McCue P.A., Shields J.A., Shields C.L., Yamamoto A., Berd D., et al. Immunoembolization of malignant liver tumors, including uveal melanoma, using granulocyte-macrophage colony-stimulating factor. J. Clin. Oncol. 2008;26:5436–5442. doi: 10.1200/JCO.2008.16.0705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Yamamoto A., Chervoneva I., Sullivan K.L., Eschelman D.J., Gonsalves C.F., Mastrangelo M.J., Berd D., Shields J.A., Shields C.L., Terai M., et al. High-dose immunoembolization: survival benefit in patients with hepatic metastases from uveal Melanoma1. Radiology. 2009;252:290–298. doi: 10.1148/radiol.2521081252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Rohaan M.W., van den Berg J.H., Kvistborg P., Haanen J.B.A.G. Adoptive transfer of tumor-infiltrating lymphocytes in melanoma: a viable treatment option. J. Immunother. Cancer. 2018;6:102. doi: 10.1186/s40425-018-0391-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Dudley M.E., Wunderlich J.R., Yang J.C., Hwu P., Schwartzentruber D.J., Topalian S.L., Sherry R.M., Marincola F.M., Leitman S.F., Seipp C.A., et al. A phase I study of nonmyeloablative chemotherapy and adoptive transfer of autologous tumor antigen-specific T lymphocytes in patients with metastatic melanoma. J. Immunother. 2002;25:243–251. doi: 10.1097/01.CJI.0000016820.36510.89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Boyiadzis M.M., Dhodapkar M.V., Brentjens R.J., Kochenderfer J.N., Neelapu S.S., Maus M.V., Porter D.L., Maloney D.G., Grupp S.A., Mackall C.L., et al. Chimeric antigen receptor (CAR) T therapies for the treatment of hematologic malignancies: clinical perspective and significance. J. Immunother. Cancer. 2018;6:137. doi: 10.1186/s40425-018-0460-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Gattinoni L., Finkelstein S.E., Klebanoff C.A., Antony P.A., Palmer D.C., Spiess P.J., Hwang L.N., Yu Z., Wrzesinski C., Heimann D.M., et al. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J. Exp. Med. 2005;202:907–912. doi: 10.1084/jem.20050732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Klebanoff C.A., Khong H.T., Antony P.A., Palmer D.C., Restifo N.P. Sinks, suppressors and antigen presenters: how lymphodepletion enhances T cell-mediated tumor immunotherapy. Trends Immunol. 2005;26:111–117. doi: 10.1016/j.it.2004.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Lowe K.L., Mackall C.L., Norry E., Amado R., Jakobsen B.K., Binder G. Fludarabine and neurotoxicity in engineered T-cell therapy. Gene Ther. 2018;25:176–191. doi: 10.1038/s41434-018-0019-6. [DOI] [PubMed] [Google Scholar]
  • 136.Goldberg J.A., Kerr D.J., Willmott N., McKillop J.H., McArdle C.S. Pharmacokinetics and pharmacodynamics of locoregional 5 fluorouracil (5FU) in advanced colorectal liver metastases. Br. J. Cancer. 1988;57:186–189. doi: 10.1038/bjc.1988.39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Cremonesi M., Ferrari M., Bodei L., Tosi G., Paganelli G. Systemic and locoregional dosimetry in receptor radionuclide therapy with peptides. Q. J. Nucl. Med. Mol. Imaging. 2006;50:288–295. [PubMed] [Google Scholar]
  • 138.Dookeran K.A., Brumfield A., Rubin J.T. A comparison of locoregional depot and systemic preparations of 9-aminocamptothecin for treatment of liver metastases in a rat tumor model: superior antitumor activity of sustained-release preparation. Ann. Surg. Oncol. 1997;4:355–360. doi: 10.1007/BF02303587. [DOI] [PubMed] [Google Scholar]
  • 139.Budker V.G., Monahan S.D., Subbotin V.M. Loco-regional cancer drug therapy: present approaches and rapidly reversible hydrophobization (RRH) of therapeutic agents as the future direction. Drug Discov. Today. 2014;19:1855–1870. doi: 10.1016/j.drudis.2014.08.009. [DOI] [PubMed] [Google Scholar]
  • 140.Allard M.-A., Malka D. Place of hepatic intra-arterial chemotherapy in the treatment of colorectal liver metastases. J. Visc. Surg. 2014;151:S21–S24. doi: 10.1016/j.jviscsurg.2013.12.003. [DOI] [PubMed] [Google Scholar]
  • 141.Kansagra A.J., Frey N.V., Bar M., Laetsch T.W., Carpenter P.A., Savani B.N., Heslop H.E., Bollard C.M., Komanduri K.V., Gastineau D.A., et al. Clinical utilization of chimeric antigen receptor T cells in B cell acute lymphoblastic leukemia: an expert opinion from the European Society for blood and marrow transplantation and the American Society for blood and marrow transplantation. Biol. Blood Marrow Transpl. 2019;25:e76–e85. doi: 10.1016/j.bbmt.2018.12.068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Brown C.E., Adusumilli P.S. Next frontiers in CAR T-cell therapy. Mol. Ther. Oncolytics. 2016;3:16028. doi: 10.1038/mto.2016.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Gattinoni L., Klebanoff C.A., Palmer D.C., Wrzesinski C., Kerstann K., Yu Z., Finkelstein S.E., Theoret M.R., Rosenberg S.A., Restifo N.P. Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells. J. Clin. Invest. 2005;115:1616–1626. doi: 10.1172/JCI24480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.McLellan A.D., Rad S.M.A.H. Chimeric antigen receptor T cell persistence and memory cell formation. Immunol. Cell Biol. 2019;97:664–674. doi: 10.1111/imcb.12254. [DOI] [PubMed] [Google Scholar]
  • 145.Turtle C.J., Hanafi L.-A., Berger C., Hudecek M., Pender B., Robinson E., Hawkins R., Chaney C., Cherian S., Chen X., et al. Immunotherapy of non-Hodgkin lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Sci. Transl. Med. 2016;8:355ra116. doi: 10.1126/scitranslmed.aaf8621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Zhang Q., Wang H., Li H., Xu J., Tian K., Yang J., Lu Z., Zheng J. Chimeric antigen receptor-modified T Cells inhibit the growth and metastases of established tissue factor-positive tumors in NOG mice. Oncotarget. 2016;8:9488–9499. doi: 10.18632/oncotarget.14367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Fisher R., Pusztai L., Swanton C. Cancer heterogeneity: implications for targeted therapeutics. Br. J. Cancer. 2013;108:479–485. doi: 10.1038/bjc.2012.581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Frenk N.E., Spring L., Muzikansky A., Vadvala H.V., Gurski J.M., Henderson L.E., Mino-Kenudson M., Ly A., Bardia A., Finkelstein D., et al. High-content biopsies facilitate molecular analyses and do not increase complication rates in patients with advanced solid tumors. JCO Precision Oncol. 2017;9:1–9. doi: 10.1200/PO.17.00081. [DOI] [PubMed] [Google Scholar]

Articles from Molecular Therapy Oncolytics are provided here courtesy of American Society of Gene & Cell Therapy

RESOURCES