- Review
- Open access
- Published:
Cancer immunotherapy and its facilitation by nanomedicine
Biomarker Research volume 12, Article number: 77 (2024)
Abstract
Cancer immunotherapy has sparked a wave of cancer research, driven by recent successful proof-of-concept clinical trials. However, barriers are emerging during its rapid development, including broad adverse effects, a lack of reliable biomarkers, tumor relapses, and drug resistance. Integration of nanomedicine may ameliorate current cancer immunotherapy. Ultra-large surface-to-volume ratio, extremely small size, and easy modification surface of nanoparticles enable them to selectively detect cells and kill cancer cells in vivo. Exciting synergistic applications of the two approaches have emerged in treating various cancers at the intersection of cancer immunotherapy and cancer nanomedicine, indicating the potential that the combination of these two therapeutic modalities can lead to new paradigms in the treatment of cancer. This review discusses the status of current immunotherapy and explores the possible opportunities that the nanomedicine platform can make cancer immunotherapy more powerful and precise by synergizing the two approaches.
Introduction
Despite significant efforts to develop quality therapies aimed at eliminating cancer, it still remains the second leading cause of death worldwide over the past decades [1, 2]. Clinical data suggest two major challenges to be overcome in cancer therapy: (1) the metastasis or spread of cancer cells [3, 4] and (2) the recurrence of dormant cells [5]. It has been recognized that immune cells play a pivotal role in controlling tumor growth, invasion, and metastasis [6, 7]. Importantly, emerging evidence suggests that each step of tumor development, from initiation through metastatic spread, involves the communication between tumor and immune cells via the secretion of cytokines and growth factors [8, 9].
To combat malignant cancer, it is undoubtedly that the immune system plays an important role. The immune system can be broadly divided into two categories: (1) the innate immune system, mainly composed of epithelial barriers, mucous membrane, monocytes and macrophages, granulocytes, neutrophils, dendritic cells (DCs), and natural killer (NK) cells [10]; and (2) the adaptive immune system, whose main components are humoral immunity dominated by B cells and cellular immunity dominated by T cells [10, 11]. In the tumor microenvironment (TME), activation of effective anti-tumor immunity requires the simultaneous engagement of the innate and adaptive immune systems. The specific execution processes summarized in Fig. 1, briefly: (1)-(2) Immature DCs phagocytose tumor-derived antigens (Ags) or recognize antigens present on tumor cells, subsequently inducing their maturation. In addition, macrophages directly engulf tumor cells via phagocytosis [12]; (3)-(4) mature DCs migrate to the lymph nodes through lymphatic vessels to active naïve T and NK cells [13, 14], (5)-(6) activated T and NK cells enter the TME through blood vessel [15, 16]; (7)-(9) in the TME, activated T and NK cells or macrophages mediated tumor lyse. However, in the TME, NK and T cells will be gradually exhausted, and exhausted T and NK cells promote tumor immune escape [17, 18].
Cancer immunotherapy, as a biological therapy, is a type of treatment that mainly takes advantages of the artificially stimulated innate and adaptive immune system to treat cancer by affecting immune cells instead of cancer cells in the above execution processes [19]. After years of developments since the first study by William Coley in 1891, various hematological and solid tumors can be treated by cancer immunotherapeutics [20, 21]. However, cancer immunotherapy still faces substantial challenges. For instance, mechanistic insights (molecular or cellular) into regulating genetic mutations, the infiltration of immune cells into the tumor bed, and the formation of immune tolerance remain elusive [22,23,24]. The treatment effects differ from patient-to-patient and cancer-to-cancer [25]. Devastatingly, it’s difficult to effectively and safely control the immune response following stimulation [26]. Moreover, the treatment expenses of current primary cancer immunotherapy can be unaffordable for most cancer patients. For example, CAR-T cell therapy is very expensive, it may cost up to $500,000 USD for a patient per year [27, 28].
To overcome these barriers, nanotechnology is being applied in the field of cancer immunotherapy by its physical (e.g., size, shape, and mechanical) and surface properties. The technology has the potential to improve the possibility of designing intelligent agents that can selectively detect cells and kill cancer cells in vivo [29, 30]. Nanomedicine, as defined by the National Institutes of Health (NIH), constitutes a branch of nanotechnology that pertains to exceedingly precise medical interventions at the molecular scale, aimed at curing diseases or restoring damaged tissues. This research field has been largely driven over the past decades and now emerges as a powerful platform for cancer treatment, particularly for cancer imaging, diagnostic, and theragnostic [31,32,33].
In this review, with the context of immuno-oncology, we elaborate on how cancer nanomedicine, an intersection field of cancer nanotechnology and cancer therapy, is revolutionizing cancer immunotherapy. The first objective is to make a brief overview of the current representative cancer immunotherapy. The second objective is to introduce the development of the cancer nanomedicine platform and how it revalorizes current cancer immunotherapy. The third objective is to provide knowledge of current clinical stage of nanomedicine for cancer immunotherapy. The final objective is to summarize the current challenges of clinical translation.
Current cancer immunotherapy
Cancer cells have developed escape mechanisms by which they co-opt both innate and adaptive immune cells to defect host immune surveillance for tumor progression. As mentioned in Fig. 1, several distinct steps must be achieved to mount effective anti-tumor immunity. Additionally, cytokines, chemokines, and the associated pathways are the main intermediaries involved in the crosstalk between cancer cells and host cells, which underlies the adjustment of tumor evasion. Thus, it’s necessary to focus on the exploration of their mechanisms to generate and regulate anti-tumor immunity [34, 35]. According to the descriptions above, we can interevent tumor evasion from at least three aspects by using various cytokines, chemokines, small molecule drugs, antibodies/proteins, viruses, or even cells [36,37,38], promoting the antigen presentation functions, promoting the production of active immune cells, and overcoming immune suppression in the tumor bed [19, 39].
Representative cancer immunotherapeutics
Representative immunotherapeutics can be summarized as: (1) Cancer vaccines. Cancer vaccines, designed to boost immune cells [40, 41], can be further subdivided into bacterial- and viral vector-based vaccines [42, 43], peptide and protein-based vaccines [44], cellular (whole cell and DC)-based vaccines [45], and nucleic acid-based vaccines including RNA vaccines, DNA vaccines, mRNA vaccines, and self-amplifying RNAs (saRNA) vaccines [46,47,48]. (2) Adoptive cell therapies (ACTs). ACTs involve the process of expanding the patient’s own T cells, NK cells or other cells with or without engineering, followed by infusion of the expanded cells into the patient with cancer [49, 50]. T-cell based cell therapy is the most popular ACT. T cell-based ACTs can be executed through at least three distinct T-cell methodologies. The first one is tumor-infiltrating lymphocytes (TILs)-based ACT, in which endogenous TILs are expanded ex vivo from a patient’s tumor before being infused back into the patient [50, 51].The second one is engineered T-cell receptor (TCR)-based ACT to recognize specific tumor antigens while it’s limited to major histocompatibility complex (MHC) expressing Ags [51]. The third one is chimeric antigen receptor (CAR)-based ACT, in which T cells from patients are engineered with a chimeric receptor consisting of an extracellular antigen recognition domain, a transmembrane domain, and a cytoplasmic signaling domain, making CAR-T cells lock onto and destroy the exact kinds of cancer [52]. (3) Immune checkpoint inhibitors (ICIs). ICIs are drugs that re-activate anti-tumor immunity by blocking co-inhibitory signaling pathways and promoting immune-mediated tumor cell clearance processes [53, 54]. For example, to evade immune surveillance, some cancer cells overexpress the programmed death-ligand 1 (PD-L1) on their surface. This will turn the “cytotoxicity brake” function on, resulting in the exhaustion of T cells and survival of the PD-L1 positive cancer cells [55]. Besides programmed cell death protein 1 (PD-1) and its ligand (PD-L1), the proteins cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) [56], lymphocyte activation gene 3 (LAG-3) [57, 58], and T-cell immunoglobulin mucin-3 (TIM-3) [59] are also common immune checkpoints (ICPs). (4) Monoclonal antibodies (mAbs). mAbs are large glycoproteins produced by B cells, targeting tumor cells directly while simultaneously promoting the induction of long-lasting anti-tumor immune responses [60]. Three types of mAbs are widely used to treat cancer, naked (or unconjugated) mAbs with nothing attached [61], conjugated mAbs with a chemotherapy drug or radioactive particle attached which are termed antibody–drug conjugates (ADCs) [62, 63], and bispecific mAbs with two different proteins attached at the same time [64]. Importantly, based on the type of heavy chain structure, there are many types of mABs and IgG is the most often form used in antibody therapy currently [65]. Ab scaffold plays an important role in tumor immunotherapy. A recent study showed that rabbit-derived Ab scaffold, a VL single domain antibody scaffold, can effectively connect 7-ethyl-10-hydroxycamptothecin (SN-38) drugs, and significantly inhibit the in vitro and in vivo proliferation of canine non-Hodgkin lymphoma (cNHL) cells, providing valuable theoretical support for Ab scaffold in tumor immunotherapy [66]. (5) Oncolytic virus (OVs) therapy. As one of the frontiers for cancer immunotherapy [67], OVs have a therapeutic potential due to their function of selectively replicating in and killing cancer cells, and spreading within the tumor, while not harming normal tissue [68, 69]. Moreover, it can be treated as an in situ vaccine [70] while it can be uploaded with immune modulatory transgenes [71,72,73,74,75], or even can be combined with other immunotherapeutics, such as cell therapy and chemotherapy [76, 77].
Pros and cons of cancer immunotherapy
Compared to traditional cancer treatments, surgical intervention, radiation therapy, and chemotherapy, immunotherapy drives significant effectiveness at prolonging the survival of patients with multiple steps, targets, and directions in attacking cancer cells, because immunotherapy can selectively target tumor tissues and reduce the damage of normal tissues [78, 79]. Cancer cells can establish mechanisms to escape immune surveillance, leading to metastases and recurrence, which are two vital factors causing death [3, 80, 81]. Immunotherapy is unequivocal through various methods to stimulate innate immunity and adaptive immunity, remodel the immune suppression in the TME to better tackle the targeted cancer cells, and amplify the pre-existing immunity. All these immunotherapeutics not only boost the immune response at the primary tumor site at the time of treatment, but also elicit systemic and prolonged protective effects by strengthening the surveillance and clearance function to prevent tumor metastasis and recurrence [82]. To date, cancer immunotherapy has been successfully applied in multiple advanced-stage malignancies; however, problems have arisen with its development. For example, immune toxicity and auto-immunity are increasingly recognized as serious clinical issues. Failure to address these issues stemming from prior failures could result in the reiteration of errors similar to those seen in some conventional therapies, thereby impeding the advancement and broader implementation of immunotherapy.
Cancer vaccines
Antigen-driven cancer vaccines can equilibrate the crosstalk between the tumor cells and the host immune system by enhancing pre-existing immunity. Cancer vaccines come into two types: prophylactic (preventative) and therapeutic (curative) [81]. It’s undeniable that the considerable success of prophylactic vaccines in preventing cancer of viral origin has been made. The most representative examples are hepatitis B virus (HBV) [83] and human papillomavirus (HPV) [84, 85] in preventing liver and cervical cancer. Meanwhile, the development of genomics and proteomics promotes the fast-evolving of therapeutic vaccines, making it easier to understand the nature of tumor-mediated tolerogenic and antigen presentation, and to identify viral antigens and mutated neo-antigens that are not subject to thymus-induced tolerance [86]. Although several clinical trials have been conducted to explore the anti-tumor efficacy of cancer vaccines and noticeable progressions have been made (Table 1), the widespread use of cancer vaccines is still a challenge. Reasons are included but not limited to (1) antigenic drift [86], (2) high disease burden, immune-suppressive and regulatory mechanisms undermining the efficacy of vaccines [87], and (3) suppressive immune cells, mainly including Regulatory T cells (Tregs) [88], TAMs [89] and myeloid-derived suppressor cells (MDSCs) [90, 91] that dampen the host immune responses.
ACTs
ACTs are personalized treatment strategies, and the Food and Drug Administration (FDA) has approved two types of CAR-T (CD19 CAR-T and BCMA CAR-T) cells for clinical trials [114, 115]. Besides these two, other types of immune cells also make achievements. For instance, TILs possess the advantage of being abundantly available. However, these cells often exhibit dysfunction due to their isolation from tumor tissues characterized by a high degree of non-synonymous gene mutations [116, 117]. Engineered TCR T cells have the advantage of being able to target intracellularly expressed proteins [118], but are limited by the diversity of human leukocyte antigens (HLA) present in the human population. CAR-T cells can be generated as individualized therapies in most patients but targeting is generally restricted to tumor-associated antigens that are expressed on the surface of tumor cells [119]. Recently, NK cell therapies have delivered promising results, showing encouraging efficacy and remarkable safety. Our group has conducted extensive research on NK cell-based cancer immunotherapy, which confirms that NK cell therapy has allogeneic potential and can be a powerful anti-cancer weapon [75, 120, 121]. Using a model with engrafted human hematopoietic cells (hHCs) in an immune-deficient mouse model, a previous preclinical study indicated that CD123 CAR-NK cells are safe (5-day OS: 100%) but not for CD123 CAR-T cells (5-day OS: 0%), although the two types of CAR immune cells have comparable antileukemia efficacy [122,123,124]. However, some challenges also exist for NK cell-based immunotherapy, such as the short half-life in vivo and limited expansion of NK cells in vitro [125]. Macrophages are one of the most promising innate immune cells, with attractive features like their phagocytic activity, capability for antigen presentation, and flexible phenotypes; however, the limited ex vivo expansion ability compared to T and NK cells and the difficulty of genomic programming with high purity are the two obstacles to overcome [126].
ICIs
ICIs are a class of immunotherapeutics that induce T cell-mediated anti-tumor responses by selectively blocking the inhibitory checkpoint receptors subject to manipulation by cancer cells [127]. Taking co-inhibitory receptors, CTLA-4 and PD-1/PD-L1, as an example, (1) ipilimumab, also named as Yervoy, is a monoclonal anti-CTLA-4 antibody. It was demonstrated that ipilimumab can shrink solid tumors and improve the overall survival rate of patients with advanced melanoma (stage III and IV). It has also been approved for adjuvant therapy (stage III melanoma) [109]. However, the blockade of CTLA-4 is accompanied by excessive activity of T cells, which not only enhances anti-tumor immune response but can also results in serious clinical toxic side effects, such as autoimmune diseases in the digestive system, liver, skin, and the nervous system [109]. (2) Similarly, targeting PD-1/PD-L1 can result in the overly activated immune T cells [128]. PD-1/PD-L1 related drugs are mainly proven effective in malignant solid tumors, and the overall effective rate in the non-selected population is not high. Thus, despite the success of anti-CTLA-4 and anti-PD-1/PD-L1 therapies, only a fraction of patients benefit from ICIs [129, 130].
MAbs
As one of the promising strategies in cancer therapeutics, the development of mAbs will continue to progress unhindered. mAbs exhibit the ability to selectively bind to antigens, inducing cytotoxicity through neutralizing or proapoptotic mechanisms, while concurrently fostering innate immune responses [131], such as antibody-dependent cellular cytotoxicity (ADCC) [132], complement-dependent cytotoxicity (CDC) [133], and antibody-dependent cellular phagocytosis (ADCP) [134]. Despite the notable clinical successes of antibody therapy, many of the mechanisms of action and their clinical relevance remain poorly understood, and therapeutic resistance remains a major challenge. Moreover, cytokine release syndrome and tumor lysis syndrome are two insurmountable hurdles [135, 136]. To better select suitable patients for mAbs therapy, a method named quantitative systems pharmacology (QSP) has been applied in a clinical trial which aimed at exploring the anti-tumor efficacy of Yervoy in treating advanced liver cancer [137].
OVs therapy
As an emerging facet of immunotherapy, OVs represent a category of viruses, either naturally occurring or artificially engineered, capable of selective replication within cancer cells, culminating in cell lysis [76, 138]. OVs can not only be used as oncolytic agents alone but they also can be used as effective carriers of anti-cancer genes and play multiple functions simultaneously such as virotherapy and gene therapy [139]. T-vec (Talimogene laherparepvec, Imlygic), for the topical treatment of unresectable skin, subcutaneous, and lymph node lesions in patients with recurrent melanoma was the first OV drug approved by the U.S. FDA, which was approved in 2015 [140]. Compared to traditional chemotherapy and radiotherapy, OVs lyse cancer cells, without damaging normal cells, tissues, and organs [138]. Therefore, oncolytic virotherapy shows potential to achieve efficacy for various types of cancers and improve the overall survival in cancer patients, which has been considered to be a promising anti-cancer therapy [141]. Nevertheless, oncolytic virotherapy’s effectiveness primarily relies on combination therapies, with the stand-alone efficacy of oncolytic viruses being subject to variability based on factors such as patients’ immune status, tumor type, and choice of oncolytic viruses. Additionally, the current OVs also have relatively poor tumor penetration and can be quickly cleared by anti-viral responses [142].
Overall, tumor immunotherapy strategies, such as cancer vaccines (e.g., cell-based vaccines), ACTs (e.g., CAR-based-ACTs, TCR-based-ACTs), ICIs (e.g., anti-PD-1 ICIs, anti-PD-L1 ICIs), mAbs, and OVs therapy (e.g., Vaccinia virus based OVs) have implemented clinical trials and some have achieved good results. The relevant clinical trials are shown in Table 1 [92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113]
The era of nanomedicine
The ultimate goal of cancer immunotherapy is to prevent cancer cell metastasis and recurrence by stimulating the immune system, and finally achieve the purpose of treatment by eliminating cancer cells. Clearly, to achieve this purpose, an active immune system with a robust and controllable immune function is necessary. However, in most situations, “robust and controllable” is challenging. Insufficient immune responses and over-activated autoimmunity are the main dilemmas in current clinical immunotherapeutics. For example, cancer vaccines can induce activated immunity for only a specific type of cancer [143], ACTs can be rendered dysfunctional as a result of tumor-mediated immune suppression [144], ICIs can cause immune-related adverse organ damage [145], and mAb therapies can result in over immune response like cytokine release syndrome [146]. Additionally, pathological and physiological barriers in our body can impede the access of immunotherapeutic drugs or native and engineered immune cells to cancer tissues and cells, making the delivery of cancer immunotherapeutic “drugs” much more difficult. Moreover, some kinds of tumors respond poorly to immunotherapeutics because of lacking an immunogenic microenvironment [147, 148]. These issues hamper the development and widespread use of cancer immunotherapeutics. Lots of efforts have been made to improve cancer immunotherapy, and within those strategies, nanoparticles (NPs)-based nanomedicine seems to be an attractive platform to overcome these shortcomings.
The concept of nanomedicine emerges in 2000 for enhancing early diagnosis and treatment of diseases such as cancers, diabetes, Alzheimer’s, Parkinson’s, and cardiovascular disease [149]. Recently, NIH re-defined it as described in the Introduction part. At present, nanomedicine can be divided into two categories: one is nanoparticle medicine, the nanometerization of the medicine itself, that is, the medicine is made into nanometer size by certain methods (e.g., suspensions, tablets, capsules) [150]; the other is nanocarrier medicine, the medicine loaded into nano-vehicles, the effect of nano-vehicles can better promote drug efficacy (e.g., liposomes, nanospheres, nanocapsules) [151,152,153].
As shown in Fig. 2 (left), the level of solubility, stability, and systemic toxicity are all safety indicators that should be considered for nanomedicines. Meanwhile, as demonstrated in Fig. 2 (right), some characteristics of NPs, such as ultra-small size, modifiable surface, and some special physicochemical characteristics are suitable for nanomedicine. As examples of outcomes, nanomedicine can increase the accumulation of drugs in the TME, controlled release of drugs, as well as target drug delivery, and even prevent degradation and clearance of medications in the circulation. The diversity of the nanomedicine is determined by the different types of NP which are shown in Fig. 3A: (I-II) surface modification: targeting ligands or surface chemistry; (III) composition: organic, inorganic, and carbon-based [154]; (IV) physical properties: shape or mechanical [155]. Moreover, many nanomedicines have been approved by the FDA for the corresponding clinical application [156,157,158,159]. This application field of nanomedicines mainly focuses on: therapeutic, diagnostic, imaging, or radiation theragnostic (Fig. 3 B). All types of nanomedicine platforms have made important contributions to oncology over the past several decades. For example, the biological and polymeric NPs can be adjusted as delivery tools due to their excellent biocompatibility, controlled release, and easily changed compositions and surface functions [160, 161]. PEGylated nano-liposomal Doxil® becomes the first FDA-approved nanomedicine [162]. Inorganic NPs have unique thermal, optical, magnetic, and electrical properties, and their chemical properties can include inertness, and stability, and can be easily engineered to perform specific functions, like regulating cell behaviors by regulating specific cellular signaling [163]. Inorganic and metallic nanomedicine Nanotherm® (MagForce), which allows cell uptake and introduces superparamagnetism to treat glioblastoma, was approved by the FDA in 2010 [164]. Crystalline nanomedicine takes advantage of the ultra-large surface-to-volume ratio of NPs to increase the dissolution velocity and saturation solubility. Additionally, increased saturation solubility due to the decreased nanoscale dimension results in the enhanced driving forces for diffusion-based mass transfer through biological structures (e.g. walls of the gastrointestinal tract), providing a feasible solution for the poorly soluble drugs [165, 166]. Both organic and inorganic materials can make the crystalline nanomedicine. The representative example is the first three FDA-approved nanocrystals: Rapamune®, Tricor®, and Emend®, three of which produced by an elan nano-system using milling approach. These nanomedicines are expected to be broadly used as bone graft substitutes that can overcome the solubility issues [164, 167]. The representative examples are covalent organic frameworks (COFs) and crystalline metal nanoparticles. The properties and functionalities of COFs are determined by the synthesis method, and thus, different synthesis methods can provide COFs with diverse functionalities which lead to the COFs’ widespread use [168]. For example, iodine- and ferrocene-loaded covalent organic framework (TADI-COF-Fc) can enhance radiotherapeutic efficacy in the treatment of radioresistant esophageal cancer, as iodine atoms on the COF framework can increase the production of reactive oxygen species (ROS) [169]. Crystalline gold nanoparticles (AuNPs) can induce mitochondria-mediated apoptosis and cell cycle arrest of cancer cells since they can depolarize the mitochondrial membrane of cancer cells [170].
Overall, NPs can be endowed with various functions. NPs-based nanomedicine is a good auxiliary platform to facilitate chemotherapy, radiotherapy, hyperthermia as well as cancer immunotherapy [171, 172].
Nanomedicine platform for cancer immunotherapy
How does nanomedicine platform assist cancer immunotherapy? How does the nanomedicine platform revolute cancer immunotherapy? To answer these two questions, we need to know the nature of cancer immunotherapy. All types of cancer immunotherapies, in nature, are “drugs”. Solubility, stability, and toxicity are parameters that need to be considered when going to clinical trials. Accumulation, release, delivery, degradation, and clearance are factors that need to be considered when mentioning drug efficiency. The limitations of cancer immunotherapies are summarized in the “Pros and cons of cancer immunotherapy” part.
To address the limitations associated with current immunotherapy, numerous nanomedicines have been developed, leveraging their advantageous attributes for various aspects of immunotherapy, including drug delivery, stimulation of anti-tumor immunity, management of tumor immune evasion, and even synergistic administration of multiple immunotherapeutic agents [173,174,175]. These nanomedicines can be generated with various compositions, properties, and modified surfaces based on their treatment purpose [176]. For example, to improve the antigen uptake and presentation process, nanomedicine generated with liposome composition or surface modified with positive charge can be used to increase the efficiency of the antigen uptake process of DCs [177]. Additionally, the size of the nanomedicine plays an important role in DCs trafficking mechanisms. Manolova et al. found that only small-size NPs (< 200 nm) can be free drainage to lymph nodes and target lymph node-resident DCs, while larger NPs (> 500 nm) have to be incorporated by skin-resident DCs for transportation to lymph nodes [178]. Nanomedicine is an exciting platform to assist cancer therapy and cancer immunotherapies, and promote their rapid development and widespread. Different kinds of nanomedicine have been generated to specifically target either tumor cells or immune cells (e.g., HER2+ BC cells, DCs, TAMs). Table 2 shows the different types of preclinical nanomedicine tested for cancer therapy or cancer immunotherapy [179,180,181,182,183,184].
Delivery vehicle for immunological agents
Precision “drug” delivery is generally a problem faced by immunotherapies, especially vaccines, mAbs, and OVs. Due to the nature of our innate immune system, mucous membrane protection, epithelial and endothelial barriers, and the phagocytosis of the phagocytic cells, the agent delivery efficiency usually be compromised, resulting in modest or even weak treatment efficacy [185, 186]. Nanomedicine is the optimal option to facilitate immunotherapies to overcome these barriers due to their nanoscale dimension and ultra-large surface-to-volume ratio, which increases solubility and enhances bio-availability [187]. This is also known as an inherent advantage of NPs, the enhanced permeability and retention (EPR) effect. A study conducted by Huang et al. shows that ultrasmall AuNPs smaller than nanometer (nm) can penetrate and localize to cancer cells better than nanoparticles larger than 10 nm. In their study, quantitative inductively coupled plasma mass spectrometry (ICP-MS) measurement shows cells uptake more AuNPs with a 2 nm diameter per cell than AuNPs with 6 nm or 15 nm diameter [188]. The high renal clearance is the other benefit of the ultra-small size nanomedicine because it can reduce the nonspecific background uptake of NPs in major organs which improves tumor-specific imaging and potential toxicity. The polymeric NPs (< 10 nm) can improve EPR-based tumor targeting and efficient renal clearance [188]. As for the delivery vehicle, the excellent bio-compatibility performance makes the liposome to be a suitable system for drug delivery [189]. In a previous study, the doxorubicin (DOX) and DOX-loaded liposomes were used to co-culture with MCF-7 cells for 24 h separately, then the IC50 of DOX in each group was measured, and the result shows that the IC50 of DOX encapsulated into liposomes was lower than that of free DOX in direct contact with cells, which means that DOX-loaded liposomes are easier to deliver DOX into cells and may have stronger anti-tumor effects [190]. Moreover, polymer, self-assembled materials, micelle, and microneedles are also optional candidates for delivery vehicles [191]. Except for delivery, accumulation, and controlled release the cargo in the target cell/tissue are the other two auxiliary functions of the nanomedicine platform that can facilitate cancer immunotherapy [192, 193]. This is mainly due to their size difference, modified surface chemistry, and specific ligand targeting design. Notably, the inner of the tumor is a microenvironment with hypoxia, low pH, and abnormal expression of glutathione and enzymes [194, 195], nanomedicine could be designed as a parameter-driven drug, such as hypoxia-, enzyme- or pH-driven drug release[196]. It can be seen in a study exploring effective treatments for osteosarcoma. In this study, calcium phosphonate was used to design pH-responsive nanomedicine termed CpG- MTX@BSA-CaZol, and this nanomedicine tends to accumulate in a low-pH tumor-associated microenvironment instead of a normal tissue [197]. Nanomedicine platforms can further be light-triggered, electric pulse-sensitive, or magnetic field-navigated with intended external conditions based on their surface chemistry conditions as Figs. 2 and 3 indicated [196, 198, 199]. Importantly, the bio-compatibility and target group modification properties make the nanomedicine excellent in protecting the antigens from premature proteolytic degradation, facilitating antigen uptake and processing by antigen-presenting cells, and decreasing toxicity to the healthy cells [200,201,202].
Antigen degradation and inefficient antigen presentation are two major shortcomings of traditional cancer vaccines [203, 204]. Kranz et al. developed a universally applicable vaccine class for systemic DC targeting and synchronized induction of both highly potent adaptive as well as type I IFN-mediated innate immune mechanisms for cancer immunotherapy by optimizing the well-known lipid carriers – the intravenously administered RNA-lipoplexes (RNA-LPX). Interestingly, the LPX can protect RNA from extracellular ribonucleases and mediates its efficient uptake and expression of the encoded antigen by DCs and macrophages in various lymphoid compartments. Additionally, RNA-LPX can precisely and effectively target the DCs and macrophages by adjusting the net charge of the particles in the best way possible without adding molecular ligands to the particles. Strikingly, they also show that RNA-LPX encoding viral or mutant neo-antigens or endogenous self-antigens induce strong effector and memory T-cell responses, and mediate potent interferon-α (IFNα)-dependent rejection of progressive tumors [205].
Nonspecific delivery of mAb therapies has the potential to induce systemic toxicity, like cytokine release syndrome and tumor lysis syndrome [146, 206]. Zhang et al. prepared an electrostatically adsorbed trastuzumab (Tmab)-bearing PLGA/PEI/lipid nanoparticles (eTmab-PPLNs), this nanomedicine includes two parts: (1) The docetaxel (DTX)-loaded PLGA/PEI/lipid hydrophobic core, composed of poly (D, L-lactide-co-glycolide), PLGA; polyethyleneimine (PEI); and lipids. (2) Electrostatically adsorbed Tmab on the surface of PLGA/PEI/lipid core as a ligand, which can target human epidermal growth factor receptor 2 (HER2)-positive breast cancer cells. The results show that the eTmab-PPLNs as a type of polymeric/lipid nanomedicine, can target the delivery of the anti-cancer drug, enhance anticancer activity, especially, decrease toxicity towards healthy cells, and reduce chemotherapeutic dose required for treatment compared to the traditional DTX [207].
Similarly, intravenous delivery of OVs is promising in cancer treatment; however, fast clearance of OVs and the severe cytokine release syndrome impede its wide application. Huang et al. for the first time, used erythrocyte lipid hybrid membrane vesicle (erythroliposome) to fully encapsulate OVs for their intravenous delivery by adding artificial membranes to cell membranes. Their results show that the fluidity of the membranes is reduced, resulting in an enhanced shielding effect on OV antigens, and consequently, the OVs display less toxicity and slower clearance after intravenous infusion, and longer circulation time. Their results also indicate that this erythroliposome encapsulated OVs markedly enhanced oncolytic efficacy to metastatic and refractory tumors [208].
Overall, nanomedicine with good biocompatibility has made noteworthy contributions to drug targeting delivery and accumulation, enhanced drug stability and bioavailability, protected drugs from degradation, prolonged their half-life, importantly, and decreased their toxicity side effects [209,210,211]. Nanomedicine greatly compensates for the deficiency of immunotherapeutics (Fig. 4).
Nanomedicine mediated ACT activity promotion
ACT, also known as cellular immunotherapy, is a form of treatment that uses the immune cells to eliminate cancer [49]. T cell, NK cell, macrophage cell are three representative ACTs in the current immunotherapy area [212, 213]. The future of ACT seems to be promising. However, despite these successes, challenges still remain. The high cost, time-consuming, and insufficient endogenous cell expansion due to the MHC restriction are obstacles that need to be overcome.
Nanomedicine in NK cell therapy
NK cell therapy recognizes their targets in an HLA-unrestricted manner and doesn’t present the risk of graft-versus-host disease (GVHD) compared to T cell [214]; however, the diversity of the NK cells and numerous regulation mechanisms, making the efficacy of NK-mediated ACT difficult to control [215]. Moreover, limited in vivo persistence and infiltration efficiency to solid tumors are other challenges to overcome [216, 217].
Attractive by nanomedicine, researchers apply it into in the field of NK therapy to improve the NK cells treatment efficacy. Wu et al. use tumor cell membranes (TM)-coated nanomedicine to stimulate resting NK cells, and the results showed that TM-loading prompted the maturation and cytotoxicity of NK cells which potentially enhanced the effect of NK cell therapy [218]. Wei et al. developed a self-assembled selenopeptide nanomedicine that successfully achieves controlled transportation and synergistic responses, addressing two critical aspects when applying nanotechnology to the field of immunotherapy [219]. This system involves three parts: delivery vesicle selenopeptide (SeP), drug like DOX, and NK immune cell. Three characteristics of SeP, a tumor-targeting motif, an enzyme-responsive cleavable linker, and an alkyl chain modified selenoamino acid tail, endow SeP with advantages of enzyme-induced size-reduction and the ROS-driven deselenization. Enzyme-induced size-reduction can enhance the drug accumulation and penetration in tumor tissue, and meanwhile, the unique ROS-driven deselenization led to the downregulation of HLA-E expression in tumor cells, which could be further facilitated by DOX, suggesting the potential of on-demand activation of NK cell-mediated immunotherapy. In conclusion, the DOX-encapsulated selenopeptide nanomedicine system (SeP/DOX) can deliver therapeutics to tumor tissue, control the release of drugs, and further activate the NK cells in a programmed manner [219] (Fig. 5A).
Nanomedicine in macrophage-mediated immunotherapy
As for macrophages, their multiple functions (maintaining homeostasis, phagocytosis, auxiliary cells to maintain T-cell tolerance, carry out surveillance for tissue integrity, maintain tissue turnover and recruit the immune system to overcome larger tissue damage, etc.), wide distributions (all organs), flexible phenotypes (M1: pro-inflammatory or tumor-suppressive, M2: anti-inflammatory or tumor-promoting), and strong plasticity making them an attractive candidate for cellular immunotherapy [220]. However, their limited ex vivo expansion ability, polarized phenotype, and the lack of safe and efficient approaches for their reprogramming greatly hindered their development and application [126].
Nanomedicine holds the ability in repolarization of tumor-promoting to tumor-suppressive macrophages to effectively activate macrophages to “eat” tumor cells. Huang et al. generated an AuPpH330/sgCD47 nanocomplexes which are PEI-coated Au nanorods with an inherent capability to induce calreticulin (CRT) exposure and electrostatically adsorbed the clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9 plasmid pH330/sgCD47. The AuPpH330/sgCD47 nanocomplexes can: (1) provide an “eat me” signal to promote the macrophage phagocytosis property, and (2) increase tumor-promoting to tumor-suppressive macrophage repolarization [221]. Ke et al. synthesized porous hollow iron oxide nanoparticles (PHNPs) for loading a PI3K inhibitor and further modified by mannose (MA) to target TAMs. The MA-NP showed good efficiency in targeting TAM, polarizing TAM to tumor-suppressive type, thereby inhibiting tumor progression [222]. In conclusion, the application of nanomedicine in macrophages is a possible strategy for macrophage-mediated immunotherapy (Fig. 5B).
Nanomedicine in adoptive T cell therapy
T cell-based ACTs, including TIL-based ACT, TCR-based ACT, and CAR-based ACT, have emerged as revolutionary immunotherapeutics for treating cancer [223]. The success of numerous clinical trials indicates its immense promise in some hematologic malignancies. However, its efficacy in solid tumors is not promising yet due to intrinsic limitations. This is exemplified by treating brain tumor in the central nervous system (CNS) with T cell therapy with the limitations of: (1) the difficulty in intertumoral delivery across anatomical niches, (2) suboptimal T cell specificity or activation, and (3) intertumoral T cell dysfunction due to the immune-suppressive TME [224].
Nanomedicine platforms may offer several advantages to overcome these limitations, as they can be designed to: (1) robustly and specifically activate or engineer T cells ex vivo, (2) encapsulate T cell-stimulating agents for positioning stimulus and controlled release, even (3) be sequentially conjugated onto T cells for added functionality without disrupting the engineering processes for TCR-/CAR-T therapies. For CNS tumors, T cells conjugated with magnetic NPs can be easily delivered into CNS solid tumors by magnetically guided. Similarly, T cells combined with photothermal-mediated NPs can promote solid tumor infiltration efficiency and enhance the cytotoxicity of T cells on CNS solid tumors [225]. Our lab conformally encapsulated donor T cells within a biocompatible and biodegradable porous film (∼450 nm in thickness) of chitosan and alginate, an approach resulting in attenuating GVHD without compromising graft-versus-leukemia (GVL) effects [226].
Genetic modification of immune cells is one of the most powerful methods to improve their tumor cell-killing efficiency, especially, CAR engineering. CD19-CAR-T and BCMA-CAR-T cell therapies are approved by the FDA for the clinical treatment of cancers [227]. CAR-viral transduction is the typical gene delivery technology with high transduction efficiency. However, the high cost, safety concerns, and small cargo capacity are the challenges that hindered its development. A nanomedicine platform can decrease the cytotoxicity and cost of the gene editing strategies by taking advantages of their composition, physical properties, as well as surface characteristics. More recently, Billingsley et al. designed an ex vivo mRNA delivery platform, C14-4/DOPE/Chol/PEG-lipid, by using ionizable lipid nanoparticles (LNPs) that can produce high quality of the CD19-CAR-T cells and elicit potent cancer-killing activity in Nalm-6 acute lymphoblastic leukemia cells [228]. Interestingly, Smith et al. designed an in situ CAR gene introducing NP which can program circulating T cells efficiently with a long-term lifespan. The design of the DNA-carrying NP includes: (1) The core cargo part which contains the microtubule-associated sequences (MTAS) and nuclear localization signals (NLS) modified poly (beta-amino ester) polymer and plasmid DNA encoding the leukemia-specific 194-1BBz CAR; (2) The T cell-targeting delivery shell part which is the electrostatically adsorbed polyglutamic acid to anti-CD3e f(ab’)2 fragments. This NP was demonstrated to be taken up by circulating T cells and importing their DNA cargo into the cell nucleus, and thus modify T cells to express leukemia-specific CARs. Additionally, the NP is easy to be manufactured and is very stable, which simplifies the long-term storage and reduces cost. Moreover, its clinical safety and efficiency were verified by using the sleeping beauty transposon system which is more suitable for patient usage compared with conventional lentiviral vectors [229]. All these above indicate the potential clinical utility of the NP-based nuclei acid delivery technique. Thus, nanomedicine is a highly imperative platform to revolute genetic editing technologies and expand the application of immunotherapy (Fig. 5C).
Nanomedicine mediated ICIs therapy
As a regulatory molecule, ICPs play a pivotal role in cancer immunotherapy. ICP has intricate relationships with ICI immunotherapeutics. The essence of ICI is a branch of mAbs, specifically promoting anti-tumor T-cell responses and response factors by targeting negative regulatory proteins (ICP: CTLA-4, PD-1/PD-L1) [230]. The function of the “brake” ICP is to protect healthy tissue to avoid failure while activating lymphocytes to remove pathogens by sending T cells a series of co-stimulatory or co-inhibitory signals via receptors. Co-stimulatory and co-inhibitory signals can cause activation and functional differentiation of T cells and termination and suppression of T-cell responses, respectively. As the first-line (chemo- and radio-therapy are still mainstay) therapies for various solid and liquid tumors, three types of ICIs, CTLA-4, PD-1 interact with ligand PD-L1 and PD-L2, and anti-PD-L1 antibodies, have been approved by the FDA [231], even though the therapeutic effects are limited in most tumor patients. Based on the above description, it’s not difficult to understand that the dilemma of current ICI immunotherapeutics is that excessive ICIs will easily cause adverse side effects like cytokine release syndrome or even immune-related adverse organ damage while a single ICI and low dose levels of ICIs are not enough to reverse the tumor immune-suppressive microenvironment and control tumors completely [232, 233].
Applying nanomedicine is a wise option to overcome the shortcomings of the ICI therapy. These nanomedicine-ICI immunotherapeutics allow for a controllable release of ICP-related antibodies and adjuvants, and the improved tumor infiltration ability of antitumor immune cells, such as T cells [196, 234]. For example, Zhang et al. reported a dual-locking nanomedicine (DLNM) that can deliver the CRISPR/CRISPR-associated (CRISPR/Cas) enzyme into tumor tissues with dual stimuli response control. This DLNM can restrict the activation of CRISPR/Cas13a (Cas13a was identified as an RNA-guided RNA-targeting CRISPR effector) in tumor tissues by responding to both the microenvironment pH and the ROS concentration in the TME. Meanwhile, the dual-locking structure effectively maintains its circulation stability and prevents CRISPR/Cas13a activation by inhibiting cellular uptake of the DLNM. Once the CRISPR/Cas13a system located the tumor cell, played a role in targeting, disrupted the programmed PD-L1 through gene editing, and achieved the controlled release, thereby leading to the safe and efficient activation of T-cell-mediated immunotherapy [235]. In another study, Wang et al. designed a self-degradable microneedle patch to treat melanoma which can sustainedly release anti-PD-1 in a controllable manner [236]. Moreover, Li et al. designed a nanoparticle to deliver an ICP targeting agent, CTLA-4-siRNA (siCTLA-4), into T cells both in vivo and in vitro. The results show that siCTLA-4 was delivered into CD4+ and CD8+ T cell subsets at tumor sites. Additionally, the CD8+ T cell ratio is significantly increased while the percentage of the Tregs is reduced. Thus, augmented activation and anti-tumor immune responses of the TILs were achieved [237]. In summary, this nanomedicine-associated ICIs therapy strategy has the potential to achieve combination therapy for enhancing anti-tumor efficacy (Fig. 6).
Nanomedicine mediated TME modification
The TME is an indispensable factor that affects the outcomes of cancer immunotherapy. Dense extracellular matrix (ECM), enrichment of cancer-associated fibroblasts (CAFs), abnormal blood vasculatures, hypoxia, tumor acidosis, abundant TAMs infiltration, and tumor immune tolerance microenvironment (TIM) are seven different main characteristics compared to the normal tissue environment [238, 239]. All these common characteristics contribute to the accelerated proliferation of cancer cells, the spread of the malignant cells, and devastatingly, the destruction of the basement membrane and the promoting metastasis of cancer cells [239,240,241]. The TME is highly imperative in cancer progression. Reprogramming the TME as an emerging and high-impact area has made outstanding achievements in suppressing tumor proliferation and improving therapeutic effects in recent studies. Taking advantage of the inherent traits of NPs and desired properties (surface modification), an engineered drug delivery system can greatly enlarge the tumor therapeutic window by regulating the TME normalization [242].
Nanomedicine platforms developed for the above seven TME properties emerge endlessly. For example, Goodman et al. conjugated collagenase onto the surface of polystyrene NPs for site-specific degradation of ECM proteins, resulting in a fourfold increase in the dose of NPs entering the multicellular spheroid model [243]. In another study, Zhao et al. made an outstanding contribution on the pancreatic ductal adenocarcinoma (PDAC) cancer treatment by using the polymeric micelle-based nanomedicine platform (M-CPA/PTX). As one of the deadliest cancers, the excessive desmoplastic stroma makes PDAC have ineffective results with intratumoral delivery of chemotherapy medicines, meanwhile, producing a self-protection mechanism against radiotherapy. The M-CPA/PTX platform includes three parts: (1) the delivery vesicle polymeric micelle, (2) a sonic hedgehog inhibitor, cyclopamine, that can deplete CAFs, and (3) a cytotoxic chemotherapy drug, paclitaxel, that can inhibit the proliferation of the PDAC cancer. The results demonstrated that M-CPA/PTX platform could remodel the TME via depleting CAFs (28% decrease) while increase the anti-tumor effect of paclitaxel, resulting in the significant efficacy in inhabiting PDAC cancer proliferation [244]. As for paclitaxel, another attractive example is the self-assembling paclitaxel filament (PF) hydrogel which stimulated the TAMs for local treatment of recurrent glioblastoma. The novelty of this study is the “drug-delivered-by-drug” strategy, which converts the poor water solubility PTX into a molecular hydrogelator that can be used for local delivery of αCD47. Next, this aqueous PF solutions can be directly deposited into the tumor resection cavity, enabling seamless hydrogel filling of the cavity and long-acting local release, meanwhile, stimulating the macrophage-mediated immune response for local treatment of recurrent glioblastoma [245]. Overall, although challenges remain when mentioned in transfer to clinical use, using the nanomedicine platforms to make the TME have normalized immune responses is a promising strategy to broaden the tumor therapeutic window.
In summary, nanomedicine has great potential in the field of cancer immunotherapy. In addition to the preclinical studies mentioned in Section 4 (Figs. 4, 5, and 6; also can be seen in Table 3), several clinical studies of nanomedicine in cancer immunotherapy, such as the Lipid NP-mediated mRNA-4157 vaccine, the Lipid NP-mediated V941 drug, and MSLN-CAR T cells that secrete PD-1/CTLA-4 nanoantibodies, have been conducted, with some studies achieving good results. The relevant clinical trials are included in Table 4 [246,247,248,249,250,251,252,253,254].
Clinical translation challenges of nanomedicine for cancer immunotherapy
Nanomedicines that incorporate some of the desired properties (e.g., conjugate antibody/protein/peptide on the surface) show great promise in the clinic, and more definitive results will be obtained in the near future. For example, the recombinant HER2 antigen and the AS15 adjuvant with liposome to treat metastatic breast cancer are at the Phase I/II stage [255]. However, despite the enormous progress that has been made in improving cancer immunotherapeutics by taking advantage of nanomedicine platforms, challenges are also gradually emerging and lying ahead. For example, permeability and EPR effects as foundational underpinnings for the nanomedicine platform delivery property, are thought to be the reason for nanomedicine accumulation in the solid tumor [256, 257]. Generally, most researchers believe that leaky tumor vasculature and poor lymphatic drainage are two reasons causing EPR effects [257]. However, this explanation of the EPR effect is somewhat oversimplified, as multiple biological steps in the systemic delivery of nanomedicine can influence the effect. Unfortunately, there is no clear mechanism that has been worked out yet. Moreover, little effort has been made to address the effect of EPR on nanomedicine-based cancer immunotherapy efficacy. Additionally, EPR effects in human remains largely unexplored, and our current understanding is mainly based on animal studies [258].
Another challenge that needs to be noted is the issue encountered in industrial production, where concretization involves several phases: (1) Controllable and reproducible synthesis: Determining the optimal physicochemical parameters like traditional drugs is essential for nanomedicine platform development. A great deal has been learned regarding independent factors such as controlled release [193], targeting delivery [259], or preventing degradation [260]. However, systematic parallel screening of numerous nanomedicine properties remains difficult due to the challenge of rapid, precise, and reproducible synthesis of nanomedicine libraries with unique features[261]. (2) Evaluation and screening: In vitro evaluation is important to select candidates before animal testing. However, traditional 2D culture in plate lacks the biocomplexity of natural tissues/organs and biomechanical cues like fluid flow. 3D cell culture systems (organ-on-chip or organoid) synergically work with animal models may offer a more reliable in vitro evaluation and screening system [261, 262]. (3) Scalable manufacturing: Good Manufacturing Practice (GMP) is a standard used to ensure that pharmaceutical products consistently meet predetermined quality standards. Manufacturing nanomedicine for the transition of it from preclinical to clinical development, subsequent commercialization, and beyond, especially the large and complex nanomedicine [258, 263, 264]. (4) Concerns of safety: Safety concerns should always be our priority, especially for clinical used “drugs”. The immunogenicity of both inorganic and organic nanomaterials is of concern, as these materials can interact with the host’s immune system, potentially contributing to autoimmunity or allergic reactions [265]. Also, the normal functions of nanomaterials depend heavily on their physicochemical properties, which can change significantly after they are administered and interact with biological components [266]. Thus, it can be difficult to predict in vivo performance by in vitro studies, and thus translating nanomedicine into clinical use is not straightforward. Therefore, how to introduce innovative methods to solve the above problems in the production of nanomedicines and how to improve the permeability and EPR effect of nanomedicines are challenges but provide us important opportunities in the clinical translation of nanomedicine for cancer immunotherapy.
Conclusions and Perspectives
We are witnessing a milestone inflection point in the field of cancer nanomedicine-assisted cancer immunotherapy. However, our current knowledge and research efforts in these two therapeutics are far from realizing the full potential of this combination modality. To facilitate the clinical development and application of cancer immunotherapeutics integrated with the platform of cancer nanomedicine, it is necessary to have a full understanding of how NPs interact with the immune system, the mechanistic insights into how NPs enable precise delivery and controlled release, and how NPs accumulate in the TME and enhance the immune response, among other factors. Controllability, reproducibility, and scalability are persistent challenges that any new generation of cancer treatment must confront. We need to determine whether nanomedicine itself possesses the necessary attributes for controllability, reproducibility, and scalability. Additionally, we must address whether nanomedicine can effectively contribute to overcoming these three “obstacles” in the clinical translation of immunotherapy. It’s important to approach these issues with objectivity.
In conclusion, we are rapidly gaining a much deeper insight into the challenges and opportunities presented by the combination of cancer nanomedicine and immunotherapy. This review has investigated the combinatory and complementary functions of cancer nanomedicine and immunotherapy that can promote the development and clinical translation of cancer therapeutics. We anticipate that this combination will shift the paradigm of cancer treatment.
Availability of data and materials
No datasets were generated or analysed during the current study.
Abbreviations
- DCs:
-
Dendritic cells
- NK cells:
-
Natural killer cells
- TME:
-
Tumor microenvironment
- Ags:
-
Antigens
- NIH:
-
National Institutes of Health
- ACTs:
-
Adoptive cell therapies
- TAMs:
-
Tumor-associated macrophages
- TILs:
-
Tumor-infiltrating lymphocytes
- TCR:
-
T-cell receptor
- MHC:
-
Major histocompatibility complex
- CAR:
-
Chimeric antigen receptor
- ICIs:
-
Immune checkpoint inhibitors
- PD-L1:
-
Programmed death-ligand 1
- PD-1:
-
Programmed cell death protein 1
- CTLA-4:
-
Cytotoxic T-lymphocyte-associated antigen 4
- LAG-3:
-
Lymphocyte activation gene 3
- TIM-3:
-
T-cell immunoglobulin mucin-3
- ICPs:
-
Immune checkpoints
- mAbs:
-
Monoclonal antibodies
- ADCs:
-
Antibody–drug conjugates
- cNHL:
-
Canine non-Hodgkin lymphoma
- OVs:
-
Oncolytic virus
- HBV:
-
Hepatitis B virus
- HPV:
-
Human papillomavirus
- Tregs:
-
Regulatory T cells
- MDSCs:
-
Myeloid-derived suppressor cells
- FDA:
-
Food and Drug Administration
- HLA:
-
Human leukocyte antigens
- hHCs:
-
Human hematopoietic cells
- ADCC:
-
Antibody-dependent cellular cytotoxicity
- CDC:
-
Complement-dependent cytotoxicity
- ADCP:
-
Antibody-dependent cellular phagocytosis
- QSP:
-
Quantitative systems pharmacology
- T-vec:
-
Talimogene laherparepvec
- NPs:
-
Nanoparticles
- COFs:
-
Covalent organic frameworks
- ROS:
-
Reactive oxygen species
- AuNPs:
-
Gold nanoparticles
- EPR:
-
Enhanced permeability and retention
- ICP-MS:
-
Inductively coupled plasma mass spectrometry
- DOX:
-
Doxorubicin
- RNA-LPX:
-
RNA-lipoplexes
- IFNα:
-
Interferon-α
- eTmab-PPLNs:
-
Electrostatically adsorbed Tmab-bearing PLGA/PEI/lipid nanoparticles
- DTX:
-
Docetaxel
- PEI:
-
Polyethylenimine
- HER2:
-
Human epidermal growth factor receptor 2
- GVHD:
-
Graft-versus-host disease
- TM:
-
Tumor cell membranes
- SeP:
-
Selenopeptide
- SeP/DOX:
-
DOX-encapsulated selenopeptide nanomedicine system
- CRT:
-
Calreticulin
- CRISPR:
-
Clustered regularly inter-spaced short palindromic repeat
- PHNPs:
-
Porous hollow iron oxide nanoparticles
- MA:
-
Mannose
- CNS:
-
Central nervous system
- GVL:
-
Graft-versus-leukemia
- LNPs:
-
Lipid nanoparticles
- MTAS:
-
Microtubule-associated sequences
- NLS:
-
Nuclear localization signals
- DLNM:
-
Dual-locking nanomedicine
- siCTLA-4:
-
CTLA-4-siRNA
- ECM:
-
Extracellular matrix
- CAFs:
-
Cancer-associated fibroblasts
- TIM:
-
Tumor immune tolerance microenvironment
- PDAC:
-
Pancreatic ductal adenocarcinoma
- M-CPA/PTX:
-
Micelle-based nanomedicine platform
- PF:
-
Paclitaxel filament
- GMP:
-
Good manufacturing practice
References
Global, regional, and national life expectancy, all-cause mortality, and cause-specific mortality for 249 causes of death, 1980–2015: a systematic analysis for the Global Burden of Disease Study 2015. Lancet. 2016;388(10053):1459–544.
Nagai H, Kim YH. Cancer prevention from the perspective of global cancer burden patterns. J Thorac Dis. 2017;9(3):448–51.
Zhang P, Meng J, Li Y, Yang C, Hou Y, Tang W, et al. Nanotechnology-enhanced immunotherapy for metastatic cancer. Innovation (Camb). 2021;2(4): 100174.
Tohme S, Simmons RL, Tsung A. Surgery for Cancer: A Trigger for Metastases. Cancer Res. 2017;77(7):1548–52.
Lebel S, Ozakinci G, Humphris G, Mutsaers B, Thewes B, Prins J, et al. From normal response to clinical problem: definition and clinical features of fear of cancer recurrence. Support Care Cancer. 2016;24(8):3265–8.
DeNardo DG, Johansson M, Coussens LM. Immune cells as mediators of solid tumor metastasis. Cancer Metastasis Rev. 2008;27(1):11–8.
Bayik D, Lathia JD. Cancer stem cell-immune cell crosstalk in tumour progression. Nat Rev Cancer. 2021;21(8):526–36.
Ruffell B, DeNardo DG, Affara NI, Coussens LM. Lymphocytes in cancer development: polarization towards pro-tumor immunity. Cytokine Growth Factor Rev. 2010;21(1):3–10.
Gan L, Qiu Z, Huang J, Li Y, Huang H, Xiang T, et al. Cyclooxygenase-2 in tumor-associated macrophages promotes metastatic potential of breast cancer cells through Akt pathway. Int J Biol Sci. 2016;12(12):1533–43.
Tomar N, De RK. A brief outline of the immune system. Methods Mol Biol. 2014;1184:3–12.
Iwasaki A, Medzhitov R. Regulation of adaptive immunity by the innate immune system. Science. 2010;327(5963):291–5.
Strioga M, Schijns V, Powell DJ Jr, Pasukoniene V, Dobrovolskiene N, Michalek J. Dendritic cells and their role in tumor immunosurveillance. Innate Immun. 2013;19(1):98–111.
Ferlazzo G, Münz C. NK cell compartments and their activation by dendritic cells. J Immunol. 2004;172(3):1333–9.
Bousso P. T-cell activation by dendritic cells in the lymph node: lessons from the movies. Nat Rev Immunol. 2008;8(9):675–84.
Ager A, Watson HA, Wehenkel SC, Mohammed RN. Homing to solid cancers: a vascular checkpoint in adoptive cell therapy using CAR T-cells. Biochem Soc Trans. 2016;44(2):377–85.
Okada K, Nannmark U, Vujanovic NL, Watkins S, Basse P, Herberman RB, et al. Elimination of established liver metastases by human interleukin 2-activated natural killer cells after locoregional or systemic adoptive transfer. Cancer Res. 1996;56(7):1599–608.
Pesce S, Greppi M, Tabellini G, Rampinelli F, Parolini S, Olive D, et al. Identification of a subset of human natural killer cells expressing high levels of programmed death 1: A phenotypic and functional characterization. J Allergy Clin Immunol. 2017;139(1):335-46.e3.
Dolina JS, Van Braeckel-Budimir N, Thomas GD, Salek-Ardakani S. CD8(+) T Cell Exhaustion in Cancer. Front Immunol. 2021;12: 715234.
Topalian SL, Weiner GJ, Pardoll DM. Cancer immunotherapy comes of age. J Clin Oncol. 2011;29(36):4828–36.
Coley WB II. Contribution to the Knowledge of Sarcoma. Ann Surg. 1891;14(3):199–220.
Im A, Pavletic SZ. Immunotherapy in hematologic malignancies: past, present, and future. J Hematol Oncol. 2017;10(1):94.
Zhang J, Endres S, Kobold S. Enhancing tumor T cell infiltration to enable cancer immunotherapy. Immunotherapy. 2019;11(3):201–13.
Curran EK, Godfrey J, Kline J. Mechanisms of Immune Tolerance in Leukemia and Lymphoma. Trends Immunol. 2017;38(7):513–25.
Reiter JG, Baretti M, Gerold JM, Makohon-Moore AP, Daud A, Iacobuzio-Donahue CA, et al. An analysis of genetic heterogeneity in untreated cancers. Nat Rev Cancer. 2019;19(11):639–50.
Vermaelen K. Vaccine Strategies to Improve Anti-cancer Cellular Immune Responses. Front Immunol. 2019;10:8.
Makkouk A, Weiner GJ. Cancer immunotherapy and breaking immune tolerance: new approaches to an old challenge. Cancer Res. 2015;75(1):5–10.
Prasad V, De Jesús K, Mailankody S. The high price of anticancer drugs: origins, implications, barriers, solutions. Nat Rev Clin Oncol. 2017;14(6):381–90.
Leighl NB, Nirmalakumar S, Ezeife DA, Gyawali B. An Arm and a Leg: The Rising Cost of Cancer Drugs and Impact on Access. Am Soc Clin Oncol Educ Book. 2021;41:1–12.
Sengupta S. Cancer Nanomedicine: Lessons for Immuno-Oncology. Trends Cancer. 2017;3(8):551–60.
Gowd V, Ahmad A, Tarique M, Suhail M, Zughaibi TA, Tabrez S, et al. Advancement of cancer immunotherapy using nanoparticles-based nanomedicine. Semin Cancer Biol. 2022;86(Pt 2):624–44.
Tong R, Kohane DS. New Strategies in Cancer Nanomedicine. Annu Rev Pharmacol Toxicol. 2016;56:41–57.
Sun T, Zhang YS, Pang B, Hyun DC, Yang M, Xia Y. Engineered nanoparticles for drug delivery in cancer therapy. Angew Chem Int Ed Engl. 2014;53(46):12320–64.
Kulkarni A, Rao P, Natarajan S, Goldman A, Sabbisetti VS, Khater Y, et al. Reporter nanoparticle that monitors its anticancer efficacy in real time. Proc Natl Acad Sci U S A. 2016;113(15):E2104–13.
Elinav E, Nowarski R, Thaiss CA, Hu B, Jin C, Flavell RA. Inflammation-induced cancer: crosstalk between tumours, immune cells and microorganisms. Nat Rev Cancer. 2013;13(11):759–71.
Sobocki BK, Basset CA, Bruhn-Olszewska B, Olszewski P, Szot O, Kaźmierczak-Siedlecka K, et al. Molecular Mechanisms Leading from Periodontal Disease to Cancer. Int J Mol Sci. 2022;23(2):970.
Kaufman HL, Kohlhapp FJ, Zloza A. Oncolytic viruses: a new class of immunotherapy drugs. Nat Rev Drug Discov. 2015;14(9):642–62.
Smith AJ. New horizons in therapeutic antibody discovery: opportunities and challenges versus small-molecule therapeutics. J Biomol Screen. 2015;20(4):437–53.
Weinmann H. Cancer Immunotherapy: Selected Targets and Small-Molecule Modulators. ChemMedChem. 2016;11(5):450–66.
Liu YT, Sun ZJ. Turning cold tumors into hot tumors by improving T-cell infiltration. Theranostics. 2021;11(11):5365–86.
Butterfield LH. Cancer vaccines Bmj. 2015;350.
Liu J, Fu M, Wang M, Wan D, Wei Y, Wei X. Cancer vaccines as promising immuno-therapeutics: platforms and current progress. J Hematol Oncol. 2022;15(1):28.
Morse MA, Hobeika AC, Osada T, Berglund P, Hubby B, Negri S, et al. An alphavirus vector overcomes the presence of neutralizing antibodies and elevated numbers of Tregs to induce immune responses in humans with advanced cancer. J Clin Invest. 2010;120(9):3234–41.
Toussaint B, Chauchet X, Wang Y, Polack B, Le Gouëllec A. Live-attenuated bacteria as a cancer vaccine vector. Expert Rev Vaccines. 2013;12(10):1139–54.
Oki Y, Younes A. Heat shock protein-based cancer vaccines. Expert Rev Vaccines. 2004;3(4):403–11.
Le DT, Pardoll DM, Jaffee EM. Cellular vaccine approaches. Cancer J. 2010;16(4):304–10.
Vogel FR, Sarver N. Nucleic acid vaccines. Clin Microbiol Rev. 1995;8(3):406–10.
Miao L, Zhang Y, Huang L. mRNA vaccine for cancer immunotherapy. Mol Cancer. 2021;20(1):41.
Faghfuri E, Pourfarzi F, Faghfouri AH, Abdoli Shadbad M, Hajiasgharzadeh K, Baradaran B. Recent developments of RNA-based vaccines in cancer immunotherapy. Expert Opin Biol Ther. 2021;21(2):201–18.
Bear AS, Fraietta JA, Narayan VK, O’Hara M, Haas NB. Adoptive Cellular Therapy for Solid Tumors. Am Soc Clin Oncol Educ Book. 2021;41:57–65.
Rohaan MW, Wilgenhof S, Haanen J. Adoptive cellular therapies: the current landscape. Virchows Arch. 2019;474(4):449–61.
Lai J, Mardiana S, House IG, Sek K, Henderson MA, Giuffrida L, et al. Adoptive cellular therapy with T cells expressing the dendritic cell growth factor Flt3L drives epitope spreading and antitumor immunity. Nat Immunol. 2020;21(8):914–26.
Newick K, O’Brien S, Moon E, Albelda SM. CAR T Cell Therapy for Solid Tumors. Annu Rev Med. 2017;68:139–52.
Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252–64.
Sharon E, Streicher H, Goncalves P, Chen HX. Immune checkpoint inhibitors in clinical trials. Chin J Cancer. 2014;33(9):434–44.
Guan J, Lim KS, Mekhail T, Chang CC. Programmed Death Ligand-1 (PD-L1) Expression in the Programmed Death Receptor-1 (PD-1)/PD-L1 Blockade: A Key Player Against Various Cancers. Arch Pathol Lab Med. 2017;141(6):851–61.
Salama AK, Hodi FS. Cytotoxic T-lymphocyte-associated antigen-4. Clin Cancer Res. 2011;17(14):4622–8.
Lythgoe MP, Liu DSK, Annels NE, Krell J, Frampton AE. Gene of the month: lymphocyte-activation gene 3 (LAG-3). J Clin Pathol. 2021;74(9):543–7.
Yu X, Huang X, Chen X, Liu J, Wu C, Pu Q, et al. Characterization of a novel anti-human lymphocyte activation gene 3 (LAG-3) antibody for cancer immunotherapy. MAbs. 2019;11(6):1139–48.
He Y, Cao J, Zhao C, Li X, Zhou C, Hirsch FR. TIM-3, a promising target for cancer immunotherapy. Onco Targets Ther. 2018;11:7005–9.
Buss NA, Henderson SJ, McFarlane M, Shenton JM, de Haan L. Monoclonal antibody therapeutics: history and future. Curr Opin Pharmacol. 2012;12(5):615–22.
Bayer V. An Overview of Monoclonal Antibodies. Semin Oncol Nurs. 2019;35(5): 150927.
Kimiz-Gebologlu I, Gulce-Iz S, Biray-Avci C. Monoclonal antibodies in cancer immunotherapy. Mol Biol Rep. 2018;45(6):2935–40.
Ponziani S, Di Vittorio G, Pitari G, Cimini AM, Ardini M, Gentile R, et al. Antibody-Drug Conjugates: The New Frontier of Chemotherapy. Int J Mol Sci. 2020;21(15):5510.
Kontermann RE, Brinkmann U. Bispecific antibodies. Drug Discov Today. 2015;20(7):838–47.
Reichert JM, Rosensweig CJ, Faden LB, Dewitz MC. Monoclonal antibody successes in the clinic. Nat Biotechnol. 2005;23(9):1073–8.
André AS, Dias JNR, Aguiar S, Nogueira S, Bule P, Carvalho JI, et al. Rabbit derived VL single-domains as promising scaffolds to generate antibody-drug conjugates. Sci Rep. 2023;13(1):4837.
Rahman MM, McFadden G. Oncolytic Viruses: Newest Frontier for Cancer Immunotherapy. Cancers (Basel). 2021;13(21):5452.
Chiocca EA, Rabkin SD. Oncolytic viruses and their application to cancer immunotherapy. Cancer Immunol Res. 2014;2(4):295–300.
Tian Y, Xie D, Yang L. Engineering strategies to enhance oncolytic viruses in cancer immunotherapy. Signal Transduct Target Ther. 2022;7(1):117.
Bartlett DL, Liu Z, Sathaiah M, Ravindranathan R, Guo Z, He Y, et al. Oncolytic viruses as therapeutic cancer vaccines. Mol Cancer. 2013;12(1):103.
Zhang Q, Liu F. Advances and potential pitfalls of oncolytic viruses expressing immunomodulatory transgene therapy for malignant gliomas. Cell Death Dis. 2020;11(6):485.
Tian L, Xu B, Chen Y, Li Z, Wang J, Zhang J, et al. Specific targeting of glioblastoma with an oncolytic virus expressing a cetuximab-CCL5 fusion protein via innate and adaptive immunity. Nat Cancer. 2022;3(11):1318–35.
Tian L, Xu B, Teng KY, Song M, Zhu Z, Chen Y, et al. Targeting Fc Receptor-Mediated Effects and the “Don’t Eat Me” Signal with an Oncolytic Virus Expressing an Anti-CD47 Antibody to Treat Metastatic Ovarian Cancer. Clin Cancer Res. 2022;28(1):201–14.
Xu B, Tian L, Chen J, Wang J, Ma R, Dong W, et al. An oncolytic virus expressing a full-length antibody enhances antitumor innate immune response to glioblastoma. Nat Commun. 2021;12(1):5908.
Ma R, Lu T, Li Z, Teng KY, Mansour AG, Yu M, et al. An Oncolytic Virus Expressing IL15/IL15Rα Combined with Off-the-Shelf EGFR-CAR NK Cells Targets Glioblastoma. Cancer Res. 2021;81(13):3635–48.
Su Y, Su C, Qin L. Current landscape and perspective of oncolytic viruses and their combination therapies. Transl Oncol. 2022;25: 101530.
Fu R, Qi R, Xiong H, Lei X, Jiang Y, He J, et al. Combination therapy with oncolytic virus and T cells or mRNA vaccine amplifies antitumor effects. Signal Transduct Target Ther. 2024;9(1):118.
Tan S, Li D, Zhu X. Cancer immunotherapy: Pros, cons and beyond. Biomed Pharmacother. 2020;124: 109821.
Koury J, Lucero M, Cato C, Chang L, Geiger J, Henry D, et al. Immunotherapies: Exploiting the Immune System for Cancer Treatment. J Immunol Res. 2018;2018:9585614.
Farkona S, Diamandis EP, Blasutig IM. Cancer immunotherapy: the beginning of the end of cancer? BMC Med. 2016;14:73.
Liu JK. Anti-cancer vaccines - a one-hit wonder? Yale J Biol Med. 2014;87(4):481–9.
Wang T, Wang D, Yu H, Feng B, Zhou F, Zhang H, et al. A cancer vaccine-mediated postoperative immunotherapy for recurrent and metastatic tumors. Nat Commun. 2018;9(1):1532.
Chang MH, You SL, Chen CJ, Liu CJ, Lai MW, Wu TC, et al. Long-term Effects of Hepatitis B Immunization of Infants in Preventing Liver Cancer. Gastroenterology. 2016;151(3):472-80.e1.
Roden R, Wu TC. How will HPV vaccines affect cervical cancer? Nat Rev Cancer. 2006;6(10):753–63.
Zepp F. Principles of vaccine design-Lessons from nature. Vaccine. 2010;28(Suppl 3):C14-24.
Carrat F, Flahault A. Influenza vaccine: the challenge of antigenic drift. Vaccine. 2007;25(39–40):6852–62.
Donninger H, Li C, Eaton JW, Yaddanapudi K. Cancer Vaccines: Promising Therapeutics or an Unattainable Dream. Vaccines (Basel). 2021;9(6):668.
Verma A, Mathur R, Farooque A, Kaul V, Gupta S, Dwarakanath BS. T-Regulatory Cells In Tumor Progression And Therapy. Cancer Manag Res. 2019;11:10731–47.
Xiang X, Wang J, Lu D, Xu X. Targeting tumor-associated macrophages to synergize tumor immunotherapy. Signal Transduct Target Ther. 2021;6(1):75.
Veglia F, Perego M, Gabrilovich D. Myeloid-derived suppressor cells coming of age. Nat Immunol. 2018;19(2):108–19.
Gabrilovich DI. Myeloid-Derived Suppressor Cells. Cancer Immunol Res. 2017;5(1):3–8.
Urman A, Ding Y, Wang H, Burkhart R A, He J, Jaffee E M, et al. Abstract CT134: Safety and immunologic impact of neoadjuvant/adjuvant GM-CSF-secreting allogenic pancreatic tumor cell vaccine (GVAX) combined with cyclophosphamide, pembrolizumab, and macrophage-targeting CSF1R inhibitor IMC-CS4 in pancreatic adenocarcinoma. Cancer Res. 2024;84(7):CT134-CT.
Vik-Mo EO, Nyakas M, Mikkelsen BV, Moe MC, Due-Tønnesen P, Suso EM, et al. Therapeutic vaccination against autologous cancer stem cells with mRNA-transfected dendritic cells in patients with glioblastoma. Cancer Immunol Immunother. 2013;62(9):1499–509.
Gulley JL, Borre M, Vogelzang NJ, Ng S, Agarwal N, Parker CC, et al. Phase III Trial of PROSTVAC in Asymptomatic or Minimally Symptomatic Metastatic Castration-Resistant Prostate Cancer. J Clin Oncol. 2019;37(13):1051–61.
Huh WK, Brady WE, Fracasso PM, Dizon DS, Powell MA, Monk BJ, et al. Phase II study of axalimogene filolisbac (ADXS-HPV) for platinum-refractory cervical carcinoma: An NRG oncology/gynecologic oncology group study. Gynecol Oncol. 2020;158(3):562–9.
Coffman-D’Annibale K, Myojin Y, Monge C, Xie C, Hrones DM, Wood BJ, et al. VB-111 (ofranergene obadenovec) in combination with nivolumab in patients with microsatellite stable colorectal liver metastases: a single center, single arm, phase II trial. J Immunother Cancer. 2024;12(1): e008079.
Weber JS, Carlino MS, Khattak A, Meniawy T, Ansstas G, Taylor MH, et al. Individualised neoantigen therapy mRNA-4157 (V940) plus pembrolizumab versus pembrolizumab monotherapy in resected melanoma (KEYNOTE-942): a randomised, phase 2b study. Lancet. 2024;403(10427):632–44.
Besse B, Charrier M, Lapierre V, Dansin E, Lantz O, Planchard D, et al. Dendritic cell-derived exosomes as maintenance immunotherapy after first line chemotherapy in NSCLC. Oncoimmunology. 2016;5(4): e1071008.
Dréno B, Khammari A, Fortun A, Vignard V, Saiagh S, Beauvais T, et al. Phase I/II clinical trial of adoptive cell transfer of sorted specific T cells for metastatic melanoma patients. Cancer Immunol Immunother. 2021;70(10):3015–30.
Nagarsheth NB, Norberg SM, Sinkoe AL, Adhikary S, Meyer TJ, Lack JB, et al. TCR-engineered T cells targeting E7 for patients with metastatic HPV-associated epithelial cancers. Nat Med. 2021;27(3):419–25.
Bagley SJ, Logun M, Fraietta JA, Wang X, Desai AS, Bagley LJ, et al. Intrathecal bivalent CAR T cells targeting EGFR and IL13Rα2 in recurrent glioblastoma: phase 1 trial interim results. Nat Med. 2024;30(5):1320–9.
Bao F, Wan W, He T, Qi F, Liu G, Hu K, et al. Autologous CD19-directed chimeric antigen receptor-T cell is an effective and safe treatment to refractory or relapsed diffuse large B-cell lymphoma. Cancer Gene Ther. 2019;26(7–8):248–55.
Geyer MB, Rivière I, Sénéchal B, Wang X, Wang Y, Purdon TJ, et al. Safety and tolerability of conditioning chemotherapy followed by CD19-targeted CAR T cells for relapsed/refractory CLL. JCI Insight. 2019;5(9).
Liu E, Marin D, Banerjee P, Macapinlac HA, Thompson P, Basar R, et al. Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. N Engl J Med. 2020;382(6):545–53.
Leighl NB, Hellmann MD, Hui R, Carcereny E, Felip E, Ahn MJ, et al. Pembrolizumab in patients with advanced non-small-cell lung cancer (KEYNOTE-001): 3-year results from an open-label, phase 1 study. Lancet Respir Med. 2019;7(4):347–57.
Borghaei H, Gettinger S, Vokes EE, Chow LQM, Burgio MA, de Castro CJ, et al. Five-Year Outcomes From the Randomized, Phase III Trials CheckMate 017 and 057: Nivolumab Versus Docetaxel in Previously Treated Non-Small-Cell Lung Cancer. J Clin Oncol. 2021;39(7):723–33.
Chen R, Zinzani PL, Lee HJ, Armand P, Johnson NA, Brice P, et al. Pembrolizumab in relapsed or refractory Hodgkin lymphoma: 2-year follow-up of KEYNOTE-087. Blood. 2019;134(14):1144–53.
Rosenberg JE, Hoffman-Censits J, Powles T, van der Heijden MS, Balar AV, Necchi A, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet. 2016;387(10031):1909–20.
Korman AJ, Garrett-Thomson SC, Lonberg N. The foundations of immune checkpoint blockade and the ipilimumab approval decennial. Nat Rev Drug Discov. 2022;21(7):509–28.
Hajda J, Lehmann M, Krebs O, Kieser M, Geletneky K, Jäger D, et al. A non-controlled, single arm, open label, phase II study of intravenous and intratumoral administration of ParvOryx in patients with metastatic, inoperable pancreatic cancer: ParvOryx02 protocol. BMC Cancer. 2017;17(1):576.
Marchand J-B, Semmrich M, Ziller C, Rehn M, Fend L, Holmkvist P, et al. Abstract 3567: Comprehensive preclinical studies of BT-001: An oncolytic vaccinia virus armed with Treg-depleting @CTLA4 and GM-CSF. Cancer Res. 2022;82(12):3567.
Nakao S, Arai Y, Tasaki M, Yamashita M, Murakami R, Kawase T, et al. Intratumoral expression of IL-7 and IL-12 using an oncolytic virus increases systemic sensitivity to immune checkpoint blockade. Sci Transl Med. 2020;12(526):eaax7992.
Zhang B, Huang J, Tang J, Hu S, Luo S, Luo Z, et al. Intratumoral OH2, an oncolytic herpes simplex virus 2, in patients with advanced solid tumors: a multicenter, phase I/II clinical trial. J Immunother Cancer. 2021;9(4).
Mullard A. FDA approves first CAR T therapy. Nat Rev Drug Discov. 2017;16(10):669.
Sjöstrand M, Sadelain M. Driving CARs to new places: locally produced BCMA CAR T cells to treat multiple myeloma. Haematologica. 2023;108(7):1721–3.
Lin B, Du L, Li H, Zhu X, Cui L, Li X. Tumor-infiltrating lymphocytes: Warriors fight against tumors powerfully. Biomed Pharmacother. 2020;132: 110873.
Paijens ST, Vledder A, de Bruyn M, Nijman HW. Tumor-infiltrating lymphocytes in the immunotherapy era. Cell Mol Immunol. 2021;18(4):842–59.
Ecsedi M, McAfee MS, Chapuis AG. The Anticancer Potential of T Cell Receptor-Engineered T Cells. Trends Cancer. 2021;7(1):48–56.
Brown CE, Mackall CL. CAR T cell therapy: inroads to response and resistance. Nat Rev Immunol. 2019;19(2):73–4.
Chu J, Deng Y, Benson DM, He S, Hughes T, Zhang J, et al. CS1-specific chimeric antigen receptor (CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma. Leukemia. 2014;28(4):917–27.
Ma S, Tang T, Wu X, Mansour AG, Lu T, Zhang J, et al. PDGF-D-PDGFRβ signaling enhances IL-15-mediated human natural killer cell survival. Proc Natl Acad Sci U S A. 2022;119(3): e2114134119.
Yilmaz A, Cui H, Caligiuri MA, Yu J. Chimeric antigen receptor-engineered natural killer cells for cancer immunotherapy. J Hematol Oncol. 2020;13(1):168.
Han J, Chu J, Keung Chan W, Zhang J, Wang Y, Cohen JB, et al. CAR-Engineered NK Cells Targeting Wild-Type EGFR and EGFRvIII Enhance Killing of Glioblastoma and Patient-Derived Glioblastoma Stem Cells. Sci Rep. 2015;5:11483.
Caruso S, De Angelis B, Del Bufalo F, Ciccone R, Donsante S, Volpe G, et al. Safe and effective off-the-shelf immunotherapy based on CAR.CD123-NK cells for the treatment of acute myeloid leukaemia. J Hematol Oncol. 2022;15(1):163.
Laskowski TJ, Biederstädt A, Rezvani K. Natural killer cells in antitumour adoptive cell immunotherapy. Nat Rev Cancer. 2022;22(10):557–75.
Poltavets AS, Vishnyakova PA, Elchaninov AV, Sukhikh GT, Fatkhudinov TK. Macrophage Modification Strategies for Efficient Cell Therapy. Cells. 2020;9(6):1535.
Bagchi S, Yuan R, Engleman EG. Immune Checkpoint Inhibitors for the Treatment of Cancer: Clinical Impact and Mechanisms of Response and Resistance. Annu Rev Pathol. 2021;16:223–49.
Riley JL. PD-1 signaling in primary T cells. Immunol Rev. 2009;229(1):114–25.
Johnson DB, Sullivan RJ, Menzies AM. Immune checkpoint inhibitors in challenging populations. Cancer. 2017;123(11):1904–11.
de Miguel M, Calvo E. Clinical Challenges of Immune Checkpoint Inhibitors. Cancer Cell. 2020;38(3):326–33.
Hubert P, Amigorena S. Antibody-dependent cell cytotoxicity in monoclonal antibody-mediated tumor immunotherapy. Oncoimmunology. 2012;1(1):103–5.
Iannello A, Ahmad A. Role of antibody-dependent cell-mediated cytotoxicity in the efficacy of therapeutic anti-cancer monoclonal antibodies. Cancer Metastasis Rev. 2005;24(4):487–99.
Glennie MJ, French RR, Cragg MS, Taylor RP. Mechanisms of killing by anti-CD20 monoclonal antibodies. Mol Immunol. 2007;44(16):3823–37.
Vozella F, Fazio F, Lapietra G, Petrucci MT, Martinelli G, Cerchione C. Monoclonal antibodies in multiple myeloma. Panminerva Med. 2021;63(1):21–7.
Shimabukuro-Vornhagen A, Gödel P, Subklewe M, Stemmler HJ, Schlößer HA, Schlaak M, et al. Cytokine release syndrome. J Immunother. Cancer. 2018;6(1):56.
Barbar T, Jaffer SI. Tumor Lysis Syndrome. Adv Chronic Kidney Dis. 2021;28(5):438-46.e1.
Sové RJ, Verma BK, Wang H, Ho WJ, Yarchoan M, Popel AS. Virtual clinical trials of anti-PD-1 and anti-CTLA-4 immunotherapy in advanced hepatocellular carcinoma using a quantitative systems pharmacology model. J Immunother Cancer. 2022;10(11): e005414.
Ma R, Li Z, Chiocca EA, Caligiuri MA, Yu J. The emerging field of oncolytic virus-based cancer immunotherapy. Trends Cancer. 2023;9(2):122–39.
Zheng M, Huang J, Tong A, Yang H. Oncolytic Viruses for Cancer Therapy: Barriers and Recent Advances. Mol Ther Oncolytics. 2019;15:234–47.
Greig SL. Talimogene Laherparepvec: First Global Approval. Drugs. 2016;76(1):147–54.
Harrington K, Freeman DJ, Kelly B, Harper J, Soria JC. Optimizing oncolytic virotherapy in cancer treatment. Nat Rev Drug Discov. 2019;18(9):689–706.
Aurelian L. Oncolytic virotherapy: the questions and the promise. Oncolytic Virother. 2013;2:19–29.
Ebrahimi N, Akbari M, Ghanaatian M, Roozbahani Moghaddam P, Adelian S, Borjian Boroujeni M, et al. Development of neoantigens: from identification in cancer cells to application in cancer vaccines. Expert Rev Vaccines. 2022;21(7):941–55.
Innamarato P, Pilon-Thomas S. Reactive myelopoiesis and the onset of myeloid-mediated immune suppression: Implications for adoptive cell therapy. Cell Immunol. 2021;361.
Gunturu KS, Pham TT, Shambhu S, Fisch MJ, Barron JJ, Debono D. Immune checkpoint inhibitors: immune-related adverse events, healthcare utilization, and costs among commercial and Medicare Advantage patients. Support Care Cancer. 2022;30(5):4019–26.
Bugelski PJ, Achuthanandam R, Capocasale RJ, Treacy G, Bouman-Thio E. Monoclonal antibody-induced cytokine-release syndrome. Expert Rev Clin Immunol. 2009;5(5):499–521.
Bonaventura P, Shekarian T, Alcazer V, Valladeau-Guilemond J, Valsesia-Wittmann S, Amigorena S, et al. Cold Tumors: A Therapeutic Challenge for Immunotherapy. Front Immunol. 2019;10:168.
Huang J, Yang B, Peng Y, Huang J, Wong SHD, Bian L, et al. Nanomedicine-Boosting Tumor Immunogenicity for Enhanced Immunotherapy. Adv Func Mater. 2021;31(21):2011171.
Moghimi SM, Hunter AC, Murray JC. Nanomedicine: current status and future prospects. Faseb j. 2005;19(3):311–30.
Jacob S, Nair AB, Shah J. Emerging role of nanosuspensions in drug delivery systems. Biomater Res. 2020;24:3.
Chariou PL, Ortega-Rivera OA, Steinmetz NF. Nanocarriers for the Delivery of Medical, Veterinary, and Agricultural Active Ingredients. ACS Nano. 2020;14(3):2678–701.
Sun X, Zhao P, Lin J, Chen K, Shen J. Recent advances in access to overcome cancer drug resistance by nanocarrier drug delivery system. Cancer Drug Resist. 2023;6(2):390–415.
Fu S, Chang L, Liu S, Gao T, Sang X, Zhang Z, et al. Temperature sensitive liposome based cancer nanomedicine enables tumour lymph node immune microenvironment remodelling. Nat Commun. 2023;14(1):2248.
Rochani A. Raveendran S. Theranostics: A New Holistic Approach in Nanomedicine; 2022. p. 573–624.
Baranov MV, Kumar M, Sacanna S, Thutupalli S, van den Bogaart G. Modulation of Immune Responses by Particle Size and Shape. Front Immunol. 2020;11.
Krauss AC, Gao X, Li L, Manning ML, Patel P, Fu W, et al. FDA Approval Summary: (Daunorubicin and Cytarabine) Liposome for Injection for the Treatment of Adults with High-Risk Acute Myeloid Leukemia. Clin Cancer Res. 2019;25(9):2685–90.
Bagley AF, Ludmir EB, Maitra A, Minsky BD, Li Smith G, Das P, et al. NBTXR3, a first-in-class radioenhancer for pancreatic ductal adenocarcinoma: Report of first patient experience. Clin Transl Radiat Oncol. 2022;33:66–9.
Reimer P, Balzer T. Ferucarbotran (Resovist): a new clinically approved RES-specific contrast agent for contrast-enhanced MRI of the liver: properties, clinical development, and applications. Eur Radiol. 2003;13(6):1266–76.
Huang W, He L, Ouyang J, Chen Q, Liu C, Tao W, et al. Triangle-Shaped Tellurium Nanostars Potentiate Radiotherapy by Boosting Checkpoint Blockade Immunotherapy. Matter. 2020;3(5):1725–53.
Xu M, Yim W, Zhou J, Zhou J, Jin Z, Moore C, et al. The Application of Organic Nanomaterials for Bioimaging, Drug Delivery, and Therapy: Spanning Various Domains. IEEE Nanatechnol Mag. 2021;15(4):8–28.
Chen G, Roy I, Yang C, Prasad PN. Nanochemistry and Nanomedicine for Nanoparticle-based Diagnostics and Therapy. Chem Rev. 2016;116(5):2826–85.
Barenholz Y. Doxil®–the first FDA-approved nano-drug: lessons learned. J Control Release. 2012;160(2):117–34.
Lohse SE, Murphy CJ. Applications of colloidal inorganic nanoparticles: from medicine to energy. J Am Chem Soc. 2012;134(38):15607–20.
Bobo D, Robinson KJ, Islam J, Thurecht KJ, Corrie SR. Nanoparticle-Based Medicines: A Review of FDA-Approved Materials and Clinical Trials to Date. Pharm Res. 2016;33(10):2373–87.
Jia L. Nanoparticle Formulation Increases Oral Bioavailability of Poorly Soluble Drugs: Approaches Experimental Evidences and Theory. Curr Nanosci. 2005;1(3):237–43.
Dizaj SM, Vazifehasl Z, Salatin S, Adibkia K, Javadzadeh Y. Nanosizing of drugs: Effect on dissolution rate. Res Pharm Sci. 2015;10(2):95–108.
Berkland C. Next Steps for Pharmaceutical Nanotechnology. J Pharm Innov. 2010;5(3):70–1.
Guo H, Liu Y, Fang C, Yan X, Zhang K, Gao H. The Cutting-Edge Progress in Covalent Organic Framework-Based Nanomedicine. Adv NanoBiomed Res. 2024;4(4):2300163.
Zhou LL, Guan Q, Zhou W, Kan JL, Teng K, Hu M, et al. A Multifunctional Covalent Organic Framework Nanozyme for Promoting Ferroptotic Radiotherapy against Esophageal Cancer. ACS Nano. 2023;17(20):20445–61.
Gao W, Wang W, Yao S, Wu S, Zhang H, Zhang J, et al. Highly sensitive detection of multiple tumor markers for lung cancer using gold nanoparticle probes and microarrays. Anal Chim Acta. 2017;958:77–84.
Hagan CTt, Mi Y, Knape N M, Wang A Z. Enhancing Combined Immunotherapy and Radiotherapy through Nanomedicine. Bioconjug Chem. 2020;31(12):2668–78.
Qu X, Zhou D, Lu J, Qin D, Zhou J, Liu HJ. Cancer nanomedicine in preoperative therapeutics: Nanotechnology-enabled neoadjuvant chemotherapy, radiotherapy, immunotherapy, and phototherapy. Bioact Mater. 2023;24:136–52.
Peng S, Xiao F, Chen M, Gao H. Tumor-Microenvironment-Responsive Nanomedicine for Enhanced Cancer Immunotherapy. Adv Sci (Weinh). 2022;9(1).
Li J, Burgess DJ. Nanomedicine-based drug delivery towards tumor biological and immunological microenvironment. Acta Pharm Sin B. 2020;10(11):2110–24.
Petrizzo A, Conte C, Tagliamonte M, Napolitano M, Bifulco K, Carriero V, et al. Functional characterization of biodegradable nanoparticles as antigen delivery system. J Exp Clin Cancer Res. 2015;34:114.
Alphandéry E. A discussion on existing nanomedicine regulation: Progress and pitfalls. Appl Mater Today. 2019;17:193–205.
Cruz LJ, Rueda F, Cordobilla B, Simón L, Hosta L, Albericio F, et al. Targeting nanosystems to human DCs via Fc receptor as an effective strategy to deliver antigen for immunotherapy. Mol Pharm. 2011;8(1):104–16.
Tran TH, Tran TTP, Nguyen HT, Phung CD, Jeong JH, Stenzel MH, et al. Nanoparticles for dendritic cell-based immunotherapy. Int J Pharm. 2018;542(1–2):253–65.
Munster P, Krop IE, LoRusso P, Ma C, Siegel BA, Shields AF, et al. Safety and pharmacokinetics of MM-302, a HER2-targeted antibody-liposomal doxorubicin conjugate, in patients with advanced HER2-positive breast cancer: a phase 1 dose-escalation study. Br J Cancer. 2018;119(9):1086–93.
Xing L, Shi Q, Zheng K, Shen M, Ma J, Li F, et al. Ultrasound-Mediated Microbubble Destruction (UMMD) Facilitates the Delivery of CA19-9 Targeted and Paclitaxel Loaded mPEG-PLGA-PLL Nanoparticles in Pancreatic Cancer. Theranostics. 2016;6(10):1573–87.
Ouyang Z, Gao Y, Yang R, Shen M, Shi X. Genetic Engineering of Dendritic Cells Using Partially Zwitterionic Dendrimer-Entrapped Gold Nanoparticles Boosts Efficient Tumor Immunotherapy. Biomacromol. 2022;23(3):1326–36.
Li Y, Wu H, Ji B, Qian W, Xia S, Wang L, et al. Targeted Imaging of CD206 Expressing Tumor-Associated M2-like Macrophages Using Mannose-Conjugated Antibiofouling Magnetic Iron Oxide Nanoparticles. ACS Appl Bio Mater. 2020;3(7):4335–47.
Qian Y, Qiao S, Dai Y, Xu G, Dai B, Lu L, et al. Molecular-Targeted Immunotherapeutic Strategy for Melanoma via Dual-Targeting Nanoparticles Delivering Small Interfering RNA to Tumor-Associated Macrophages. ACS Nano. 2017;11(9):9536–49.
Spinato C, Perez Ruiz de Garibay A, Kierkowicz M, Pach E, Martincic M, Klippstein R, et al. Design of antibody-functionalized carbon nanotubes filled with radioactivable metals towards a targeted anticancer therapy. Nanoscale. 2016;8(25):12626–38.
Jain K, Mehra NK, Jain NK. Nanotechnology in Drug Delivery: Safety and Toxicity Issues. Curr Pharm Des. 2015;21(29):4252–61.
Ban C, Jo M, Park YH, Kim JH, Han JY, Lee KW, et al. Enhancing the oral bioavailability of curcumin using solid lipid nanoparticles. Food Chem. 2020;302.
Irvine DJ, Dane EL. Enhancing cancer immunotherapy with nanomedicine. Nat Rev Immunol. 2020;20(5):321–34.
Huang K, Ma H, Liu J, Huo S, Kumar A, Wei T, et al. Size-dependent localization and penetration of ultrasmall gold nanoparticles in cancer cells, multicellular spheroids, and tumors in vivo. ACS Nano. 2012;6(5):4483–93.
Akbarzadeh A, Rezaei-Sadabady R, Davaran S, Joo SW, Zarghami N, Hanifehpour Y, et al. Liposome: classification, preparation, and applications. Nanoscale Res Lett. 2013;8(1):102.
Ding Y, Cui W, Sun D, Wang GL, Hei Y, Meng S, et al. In vivo study of doxorubicin-loaded cell-penetrating peptide-modified pH-sensitive liposomes: biocompatibility, bio-distribution, and pharmacodynamics in BALB/c nude mice bearing human breast tumors. Drug Des Devel Ther. 2017;11:3105–17.
Bookstaver ML, Tsai SJ, Bromberg JS, Jewell CM. Improving Vaccine and Immunotherapy Design Using Biomaterials. Trends Immunol. 2018;39(2):135–50.
Matsumura Y, Maeda H. A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res. 1986;46(12 Pt 1):6387–92.
Jin Q, Liu Z, Chen Q. Controlled release of immunotherapeutics for enhanced cancer immunotherapy after local delivery. J Control Release. 2021;329:882–93.
Augustin RC, Delgoffe GM, Najjar YG. Characteristics of the Tumor Microenvironment That Influence Immune Cell Functions: Hypoxia, Oxidative Stress, Metabolic Alterations. Cancers (Basel). 2020;12(12):3802.
Cook JA, Gius D, Wink DA, Krishna MC, Russo A, Mitchell JB. Oxidative stress, redox, and the tumor microenvironment. Semin Radiat Oncol. 2004;14(3):259–66.
Liu Q, Duo Y, Fu J, Qiu M, Sun Z, Adah D, et al. Nano-immunotherapy: Unique mechanisms of nanomaterials in synergizing cancer immunotherapy. Nano Today. 2021;36.
Wang Y, Wu Y, Li L, Ma C, Zhang S, Lin S, et al. Chemotherapy-Sensitized In Situ Vaccination for Malignant Osteosarcoma Enabled by Bioinspired Calcium Phosphonate Nanoagents. ACS Nano. 2023;17(7):6247–60.
Cheng HW, Tsao HY, Chiang CS, Chen SY. Advances in Magnetic Nanoparticle-Mediated Cancer Immune-Theranostics. Adv Healthc Mater. 2021;10(1).
Latorre A, Couleaud P, Aires A, Cortajarena AL, Somoza Á. Multifunctionalization of magnetic nanoparticles for controlled drug release: a general approach. Eur J Med Chem. 2014;82:355–62.
Fang RH, Kroll AV, Zhang L. Nanoparticle-Based Manipulation of Antigen-Presenting Cells for Cancer Immunotherapy. Small. 2015;11(41):5483–96.
Kheirollahpour M, Mehrabi M, Dounighi NM, Mohammadi M, Masoudi A. Nanoparticles and Vaccine Development. Pharm Nanotechnol. 2020;8(1):6–21.
Ahmad A, Khan F, Mishra RK, Khan R. Precision Cancer Nanotherapy: Evolving Role of Multifunctional Nanoparticles for Cancer Active Targeting. J Med Chem. 2019;62(23):10475–96.
Goyvaerts C, Breckpot K. Pros and Cons of Antigen-Presenting Cell Targeted Tumor Vaccines. J Immunol Res. 2015;2015.
He Q, Gao H, Tan D, Zhang H, Wang JZ. mRNA cancer vaccines: Advances, trends and challenges. Acta Pharm Sin B. 2022;12(7):2969–89.
Kranz LM, Diken M, Haas H, Kreiter S, Loquai C, Reuter KC, et al. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature. 2016;534(7607):396–401.
Xia S, Ma JT, Raschi E, Ma R, Zhang BK, Guo L, et al. Tumor Lysis Syndrome with CD20 Monoclonal Antibodies for Chronic Lymphocytic Leukemia: Signals from the FDA Adverse Event Reporting System. Clin Drug Investig. 2023;43(10):773–83.
Zhang X, Liu J, Li X, Li F, Lee RJ, Sun F, et al. Trastuzumab-Coated Nanoparticles Loaded With Docetaxel for Breast Cancer Therapy. Dose Response. 2019;17(3):1559325819872583.
Huang H, Sun M, Liu M, Pan S, Liu P, Cheng Z, et al. Full encapsulation of oncolytic virus using hybrid erythroctye-liposome membranes for augmented anti-refractory tumor effectiveness. Nano Today. 2022;47.
Smith MJ, Brown JM, Zamboni WC, Walker NJ. From immunotoxicity to nanotherapy: the effects of nanomaterials on the immune system. Toxicol Sci. 2014;138(2):249–55.
Kapadia CH, Perry JL, Tian S, Luft JC, DeSimone JM. Nanoparticulate immunotherapy for cancer. J Control Release. 2015;219:167–80.
Manjili HK, Malvandi H, Mousavi MS, Attari E, Danafar H. In vitro and in vivo delivery of artemisinin loaded PCL-PEG-PCL micelles and its pharmacokinetic study. Artif Cells Nanomed Biotechnol. 2018;46(5):926–36.
Pan K, Farrukh H, Chittepu V, Xu H, Pan CX, Zhu Z. CAR race to cancer immunotherapy: from CAR T, CAR NK to CAR macrophage therapy. J Exp Clin Cancer Res. 2022;41(1):119.
Shah Z, Tian L, Li Z, Jin L, Zhang J, Li Z, et al. Human anti-PSCA CAR macrophages possess potent antitumor activity against pancreatic cancer. Cell Stem Cell. 2024;31(6):803-17.e6.
Gill S, Olson JA, Negrin RS. Natural killer cells in allogeneic transplantation: effect on engraftment, graft- versus-tumor, and graft-versus-host responses. Biol Blood Marrow Transplant. 2009;15(7):765–76.
Myers JA, Miller JS. Exploring the NK cell platform for cancer immunotherapy. Nat Rev Clin Oncol. 2021;18(2):85–100.
Nayyar G, Chu Y, Cairo MS. Overcoming Resistance to Natural Killer Cell Based Immunotherapies for Solid Tumors. Front Oncol. 2019;9:51.
Peled T, Brachya G, Persi N, Lador C, Olesinski E, Landau E, et al. Enhanced In Vivo Persistence and Proliferation of NK Cells Expanded in Culture with the Small Molecule Nicotinamide: Development of a Clinical-Applicable Method for NK Expansion. Blood. 2017;130:657.
Wu D, Shou X, Zhang Y, Li Z, Wu G, Wu D, et al. Cell membrane-encapsulated magnetic nanoparticles for enhancing natural killer cell-mediated cancer immunotherapy. Nanomedicine. 2021;32.
Wei Z, Yi Y, Luo Z, Gong X, Jiang Y, Hou D, et al. Selenopeptide Nanomedicine Activates Natural Killer Cells for Enhanced Tumor Chemoimmunotherapy. Adv Mater. 2022;34(17).
Lee S, Kivimäe S, Dolor A, Szoka FC. Macrophage-based cell therapies: The long and winding road. J Control Release. 2016;240:527–40.
Huang Q, Su M, Zhao L, Zhang Z, Zhang Y, Yang X, et al. NIR-II Light-driven genetically edited nanoparticles with inherent CRT-inducing capability for macrophage-mediated immunotherapy. Nano Today. 2023;50.
Li K, Lu L, Xue C, Liu J, He Y, Zhou J, et al. Polarization of tumor-associated macrophage phenotype via porous hollow iron nanoparticles for tumor immunotherapy in vivo. Nanoscale. 2020;12(1):130–44.
Feldman SA, Assadipour Y, Kriley I, Goff SL, Rosenberg SA. Adoptive Cell Therapy–Tumor-Infiltrating Lymphocytes, T-Cell Receptors, and Chimeric Antigen Receptors. Semin Oncol. 2015;42(4):626–39.
Hwang EI, Sayour EJ, Flores CT, Grant G, Wechsler-Reya R, Hoang-Minh LB, et al. The current landscape of immunotherapy for pediatric brain tumors. Nat Cancer. 2022;3(1):11–24.
Balakrishnan PB, Sweeney EE. Nanoparticles for Enhanced Adoptive T Cell Therapies and Future Perspectives for CNS Tumors. Front Immunol. 2021;12.
Zhao S, Zhang L, Han J, Chu J, Wang H, Chen X, et al. Conformal Nanoencapsulation of Allogeneic T Cells Mitigates Graft-versus-Host Disease and Retains Graft-versus-Leukemia Activity. ACS Nano. 2016;10(6):6189–200.
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.
Billingsley MM, Singh N, Ravikumar P, Zhang R, June CH, Mitchell MJ. Ionizable Lipid Nanoparticle-Mediated mRNA Delivery for Human CAR T Cell Engineering. Nano Lett. 2020;20(3):1578–89.
Smith TT, Stephan SB, Moffett HF, McKnight LE, Ji W, Reiman D, et al. In situ programming of leukaemia-specific T cells using synthetic DNA nanocarriers. Nat Nanotechnol. 2017;12(8):813–20.
Qin S, Xu L, Yi M, Yu S, Wu K, Luo S. el immune checkpoint targets: moving beyond PD-1 and CTLA-4. Mol Cancer. 2019;18(1):155.
Hargadon KM, Johnson CE, Williams CJ. Immune checkpoint blockade therapy for cancer: An overview of FDA-approved immune checkpoint inhibitors. Int Immunopharmacol. 2018;62:29–39.
Suntharalingam G, Perry MR, Ward S, Brett SJ, Castello-Cortes A, Brunner MD, et al. Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N Engl J Med. 2006;355(10):1018–28.
Gong J, Hendifar A, Tuli R, Chuang J, Cho M, Chung V, et al. Combination systemic therapies with immune checkpoint inhibitors in pancreatic cancer: overcoming resistance to single-agent checkpoint blockade. Clin Transl Med. 2018;7(1):32.
Duan X, Chan C, Han W, Guo N, Weichselbaum RR, Lin W. Immunostimulatory nanomedicines synergize with checkpoint blockade immunotherapy to eradicate colorectal tumors. Nat Commun. 2019;10(1):1899.
Zhang Z, Wang Q, Liu Q, Zheng Y, Zheng C, Yi K, et al. Dual-Locking Nanoparticles Disrupt the PD-1/PD-L1 Pathway for Efficient Cancer Immunotherapy. Adv Mater. 2019;31(51).
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.
Li SY, Liu Y, Xu CF, Shen S, Sun R, Du XJ, et al. Restoring anti-tumor functions of T cells via nanoparticle-mediated immune checkpoint modulation. J Control Release. 2016;231:17–28.
Mistrangelo M, Allaix M E, Arezzo A, Morino M, et al. The outcome of rectal cancer after early salvage TME following TEM compared with primary TME: a case-matched study. Tech Coloproctol. 2014;18(1):81.
Yang B, Meng F, Zhang J, Chen K, Meng S, Cai K, et al. Engineered drug delivery nanosystems for tumor microenvironment normalization therapy. Nano Today. 2023;49.
Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.
Monboisse JC, Oudart JB, Ramont L, Brassart-Pasco S, Maquart FX. Matrikines from basement membrane collagens: a new anti-cancer strategy. Biochim Biophys Acta. 2014;1840(8):2589–98.
Yang S, Gao H. Nanoparticles for modulating tumor microenvironment to improve drug delivery and tumor therapy. Pharmacol Res. 2017;126:97–108.
Goodman TT, Olive PL, Pun SH. Increased nanoparticle penetration in collagenase-treated multicellular spheroids. Int J Nanomedicine. 2007;2(2):265–74.
Zhao J, Wang H, Hsiao CH, Chow DS, Koay EJ, Kang Y, et al. Simultaneous inhibition of hedgehog signaling and tumor proliferation remodels stroma and enhances pancreatic cancer therapy. Biomaterials. 2018;159:215–28.
Wang F, Huang Q, Su H, Sun M, Wang Z, Chen Z, et al. Self-assembling paclitaxel-mediated stimulation of tumor-associated macrophages for postoperative treatment of glioblastoma. Proc Natl Acad Sci U S A. 2023;120(18).
Bauman J, Burris H, Clarke J, Patel M, Cho D, Gutierrez M, et al. Safety, tolerability, and immunogenicity of mRNA-4157 in combination with pembrolizumab in subjects with unresectable solid tumors (KEYNOTE-603): an update. J Immunother Cancer. 2020;8(3):A477.
Patel M, Jimeno A, Wang D, Stemmer S, Bauer T, Sweis R, et al. Phase 1 study of mRNA-2752, a lipid nanoparticle encapsulating mRNAs encoding human OX40L/IL-23/IL-36γ, for intratumoral (ITu) injection +/- durvalumab in advanced solid tumors and lymphoma. J Immunother Cancer. 2021;9(2):A569.
Nagasaka M. Emerging Mechanisms to Target KRAS Directly. J Thorac Oncol. 2021;16(3):96–7.
Creemers JHA, Pawlitzky I, Grosios K, Gileadi U, Middleton MR, Gerritsen WR, et al. Assessing the safety, tolerability and efficacy of PLGA-based immunomodulatory nanoparticles in patients with advanced NY-ESO-1-positive cancers: a first-in-human phase I open-label dose-escalation study protocol. BMJ Open. 2021;11(11).
Xie P, Yang ST, Huang Y, Zeng C, Xin Q, Zeng G, et al. Carbon Nanoparticles-Fe(II) Complex for Efficient Tumor Inhibition with Low Toxicity by Amplifying Oxidative Stress. ACS Appl Mater Interfaces. 2020;12(26):29094–102.
Thivat E, Casile M, Moreau J, Molnar I, Dufort S, Seddik K, et al. Phase I/II study testing the combination of AGuIX nanoparticles with radiochemotherapy and concomitant temozolomide in patients with newly diagnosed glioblastoma (NANO-GBM trial protocol). BMC Cancer. 2023;23(1):344.
Farrukh H, Zhu Z, Risco J, Lam K, Pan C-x. A phase I dose-escalation clinical trial with PLZ4-coated paclitaxel-loaded micelles (PPM) in patients with recurrent or refractory non-myoinvasive bladder cancer. J Clin Oncol. 2023;41:TPS4615.
Thaker PH, Bradley WH, Leath CA, Gunderson Jackson C, Borys N, Anwer K, et al. GEN-1 in Combination with Neoadjuvant Chemotherapy for Patients with Advanced Epithelial Ovarian Cancer: A Phase I Dose-escalation Study. Clin Cancer Res. 2021;27(20):5536–45.
Sahin U, Oehm P, Derhovanessian E, Jabulowsky RA, Vormehr M, Gold M, et al. An RNA vaccine drives immunity in checkpoint-inhibitor-treated melanoma. Nature. 2020;585(7823):107–12.
Curigliano G, Romieu G, Campone M, Dorval T, Duck L, Canon JL, et al. A phase I/II trial of the safety and clinical activity of a HER2-protein based immunotherapeutic for treating women with HER2-positive metastatic breast cancer. Breast Cancer Res Treat. 2016;156(2):301–10.
Maeda H. Toward a full understanding of the EPR effect in primary and metastatic tumors as well as issues related to its heterogeneity. Adv Drug Deliv Rev. 2015;91:3–6.
Huynh E, Zheng G. Cancer nanomedicine: addressing the dark side of the enhanced permeability and retention effect. Nanomedicine (Lond). 2015;10(13):1993–5.
Shi J, Kantoff PW, Wooster R, Farokhzad OC. Cancer nanomedicine: progress, challenges and opportunities. Nat Rev Cancer. 2017;17(1):20–37.
Lin JY, Liu HJ, Wu Y, Jin JM, Zhou YD, Zhang H, et al. Targeted Protein Degradation Technology and Nanomedicine: Powerful Allies against Cancer. Small. 2023;19(18).
Klochkov SG, Neganova ME, Nikolenko VN, Chen K, Somasundaram SG, Kirkland CE, et al. Implications of nanotechnology for the treatment of cancer: Recent advances. Semin Cancer Biol. 2021;69:190–9.
Low LA, Mummery C, Berridge BR, Austin CP, Tagle DA. Organs-on-chips: into the next decade. Nat Rev Drug Discov. 2021;20(5):345–61.
Bär SI, Biersack B, Schobert R. 3D cell cultures, as a surrogate for animal models, enhance the diagnostic value of preclinical in vitro investigations by adding information on the tumour microenvironment: a comparative study of new dual-mode HDAC inhibitors. Invest New Drugs. 2022;40(5):953–61.
Zheng C, Li M, Ding J. Challenges and Opportunities of Nanomedicines in Clinical Translation. BIO Integration. 2021;2:57–60.
Wicki A, Witzigmann D, Balasubramanian V, Huwyler J. Nanomedicine in cancer therapy: challenges, opportunities, and clinical applications. J Control Release. 2015;200:138–57.
Ilinskaya AN, Dobrovolskaia MA. Understanding the immunogenicity and antigenicity of nanomaterials: Past, present and future. Toxicol Appl Pharmacol. 2016;299:70–7.
Mu Q, Jiang G, Chen L, Zhou H, Fourches D, Tropsha A, et al. Chemical basis of interactions between engineered nanoparticles and biological systems. Chem Rev. 2014;114(15):7740–81.
Acknowledgements
Figures 1- 4 are made using www.BioRender.com.
Funding
Not applicable.
Author information
Authors and Affiliations
Contributions
CS, YJ, and W X conceived the review. CS, LX and WH wrote the manuscript. All the authors approved the final version of the manuscript.
Corresponding authors
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.
About this article
Cite this article
Sui, C., Wu, H., Li, X. et al. Cancer immunotherapy and its facilitation by nanomedicine. Biomark Res 12, 77 (2024). https://doi.org/10.1186/s40364-024-00625-6
Received:
Accepted:
Published:
DOI: https://doi.org/10.1186/s40364-024-00625-6