Next Article in Journal
Complex Analysis of Endothelial Markers as Potential Prognostic Indicators in Luminal Invasive Breast Carcinoma Patients: Outcomes of a Six-Year Observational Study
Previous Article in Journal
Evaluation and Application of Silk Fibroin Based Biomaterials to Promote Cartilage Regeneration in Osteoarthritis Therapy
Previous Article in Special Issue
Dabrafenib-Trametinib and Radiotherapy for Oligoprogressive BRAF Mutant Advanced Melanoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Enhancing Immunogenicity in Metastatic Melanoma: Adjuvant Therapies to Promote the Anti-Tumor Immune Response

1
Department of Development and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
2
Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
3
Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
4
Department of Urology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
5
Institute of Onco-Physics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(8), 2245; https://doi.org/10.3390/biomedicines11082245
Submission received: 27 June 2023 / Revised: 26 July 2023 / Accepted: 4 August 2023 / Published: 10 August 2023
(This article belongs to the Special Issue Metastatic Melanoma: Current Status and Future Prospects)

Abstract

:
Advanced melanoma is an aggressive form of skin cancer characterized by low survival rates. Less than 50% of advanced melanoma patients respond to current therapies, and of those patients that do respond, many present with tumor recurrence due to resistance. The immunosuppressive tumor-immune microenvironment (TIME) remains a major obstacle in melanoma therapy. Adjuvant treatment modalities that enhance anti-tumor immune cell function are associated with improved patient response. One potential mechanism to stimulate the anti-tumor immune response is by inducing immunogenic cell death (ICD) in tumors. ICD leads to the release of damage-associated molecular patterns within the TIME, subsequently promoting antigen presentation and anti-tumor immunity. This review summarizes relevant concepts and mechanisms underlying ICD and introduces the potential of non-ablative low-intensity focused ultrasound (LOFU) as an immune-priming therapy that can be combined with ICD-inducing focal ablative therapies to promote an anti-melanoma immune response.

1. Introduction

Melanoma is a rare and aggressive form of skin cancer that is responsible for 75% of skin cancer-related deaths [1,2]. When diagnosed in advanced stages, melanoma is associated with a very poor prognosis with a patient survival rate as low as 30% [2]. Immune checkpoint inhibitors (CIs), such as anti-CTLA-4 and anti-PD-1 blocking antibodies (Table 1), target the inhibitory receptors on T-cells, including programmed cell death protein-1 (PD-1) and cytotoxic T-lymphocyte-associated protein-4 (CTLA-4), with the goal of enhancing anti-tumor T-cell function. These checkpoint inhibitors have significantly improved survival rates in patients with advanced melanoma.
CI treatment efficacy has been shown to correlate with the number of tumor-infiltrating lymphocytes, indicating that an endogenous anti-tumor T-cell immunity is associated with increased response [15]. However, CI therapies are only effective in 10–40% of patients when used as monotherapies, and approximately 53% when combined as dual therapies [16]. Even in cases where CI therapy is effective initially, many patients proceed to present with tumor recurrence. Potential mechanisms underlying this loss of CI efficacy include the selection for tumor cells that are resistant to CI therapy, the upregulation of other inhibitory receptors, and the clearance of CI antibodies by tumor-associated macrophages [17,18]. Metastatic melanoma cells have also been shown to release exosomes, extracellular vesicles that are approximately 30 to 150 nm in size, with a high expression of PD-L1 which subsequently suppresses the anti-tumor immune response [19]. Conversely, the expression of PD-L1 by myeloid-derived immune cells such as conventional dendritic cells and macrophages within the tumor microenvironment is required for the generation of a strong response to CI therapies [20,21]. Additionally, CI therapies are associated with adverse events, some of which can be severe and which include autoimmune-related disorders such as vitiligo and colitis, and even death [22]. The overall incidence of immune-related adverse events (IRAE) in advanced melanoma patients is dependent on the checkpoint inhibitor therapy regimen: approximately 23% of patients treated with pembrolizumab monotherapy experience IRAE, and this incidence increases to 62% with ipilimumab monotherapy and 83% with combined CI therapies [23]. Unfortunately, there is a positive association between CI efficacy and number of adverse events [24]. Therefore, balancing the low response rate and the risk of adverse effects is vital when determining whether a patient should receive CI therapy.
Biomarkers are a potential tool to guide personalized treatment plans for melanoma patients. The programmed death-ligand 1 (PD-L1), expressed on tumor cells, binds to T-cell surface PD-1, thereby attenuating the anti-tumor T-cell response. Although PD-L1 expression, measured by immunohistochemistry, has been approved by the Food and Drug Administration (FDA) as a biomarker to predict responsiveness to immunotherapy in certain cancers (non-small cell lung cancer, gastric/gastroesophageal junction adenocarcinoma, triple-negative breast cancer, and others), the utility of this biomarker is still under investigation [25,26]. However, such biomarkers may play an important role in directing patient therapy in the future.
A major challenge in cancer immunotherapy is the immunosuppressive tumor-immune microenvironment (TIME). The generation of a robust anti-tumor response relies on dendritic cells (DCs), professional antigen-presenting cells which must first recognize tumor antigens and damage-associated molecular patterns (DAMP) within the TIME via pattern-recognition receptors, and then process the antigens and migrate to the draining lymph node, where they present tumor antigen peptides to both CD4+ and CD8+ T-cells [27]. However, within the TIME, immunosuppressive factors impede DC differentiation and activation, leading to systemic changes in DC populations and function that prevent efficient antigen presentation [28]. As a result, melanoma patients present with decreased numbers of peripheral and intra-tumoral DCs [29,30,31,32]. Additionally, DC subpopulations in melanoma patients favor an immature phenotype and lack co-stimulatory signals [30,31,33,34]. These immature DCs can lead to the establishment of T-cell anergy, in which T-cells, the critical effectors of the TIME, enter a dysfunctional, tolerant state with notably reduced effector cytokine secretion and proliferation [35,36,37]. In melanoma, intra-tumoral populations of T-cells often show anergic and exhausted phenotypes [38], and these populations are associated with increased melanoma persistence [34].

2. Immunogenic Cell Death (ICD)

Focal therapies that induce ICD in tumor cells have the potential to overcome immunosuppression in the TIME and to improve patient responses to current immunotherapies [39]. Tumor-cell ICD triggers a local, anti-tumor immune reaction (Figure 1) that can potentially prevent resistance to CI therapies [39,40]. Typically, tumor cells that are undergoing ICD experience endoplasmic reticular (ER) stress due to oxidative stress and/or the accumulation of misfolded proteins in the ER, with subsequent activation of the unfolded protein response (UPR) [39,40]. This leads to the translocation of calreticulin from the endoplasmic reticulum (ER) to the plasma membrane (ecto-CRT) [39,40,41]. Ecto-CRT promotes the phagocytosis of tumor cells by DCs [42]. Additionally, ecto-CRT also promotes natural killer cell-mediated tumor cell death [43]. Studies have shown that ecto-CRT is essential for the induction of ICD, since melanoma cells that are unable to translocate CRT cannot stimulate ICD and are subsequently unable to induce an anti-tumor immune response [41]. The cofactors required for the translocation of calreticulin are also involved in other cellular processes such as autophagy that have also been implicated in pro-immunogenic signaling within the TIME.
ICD, immunogenic cell death; ER, endoplasmic reticulum; UPR, unfolded protein response; PERK, protein kinase RNA-like ER kinase; ATF4, activating transcription factor 4; DAMP, damage-associated molecular pattern; HMGB1, high-mobility group box protein 1; ATP, adenosine triphosphate (Created with BioRender.com).
Autophagy, a lysosome-dependent process of cytosolic content degradation, plays important roles in both immune evasion [44,45,46] and tumor growth [47,48,49]. Recent studies have shown that inducing autophagy in melanoma cells can protect them from immediate apoptosis and that this delay in apoptosis is necessary for the generation of pro-immunogenic signaling and ICD markers [50]. Increased levels of autophagy in tumor cells prior to ICD is required for the secretion of adenosine triphosphate (ATP), which acts as a damage-associated molecular pattern (DAMP) and chemoattractant to enhance the migration of inflammatory monocytes and DCs to the tumor site. Cancer cell lines depleted of autophagy-specific machinery are not able to activate a tumor-specific immune response [51,52]. Although autophagy-deficient cells can undergo apoptosis and necrosis, they show a reduction in secreted ATP, which is required to enhance the recruitment of immune cells to the tumor site [51]. Increased levels of autophagy within tumors has been shown to be positively correlated with improved patient prognosis [52].
As tumor cells undergo ICD, the dying cells also release high-mobility group box protein 1 (HMGB1). HMGB1 is normally found within the nucleus, but as cells are damaged, it can translocate to the cytosol and be secreted into the extracellular space, where it acts as a DAMP that subsequently activates the immune response [53]. HMGB1 directly and indirectly interacts with a variety of receptors including toll-like receptor 4 (TLR4) and the receptor for advanced glycation end-products (RAGE) [48]. HMGB1 binding with TLR4 on antigen-presenting cells leads to the direct induction of an adaptive anti-tumor immune response, while its binding with RAGE induces autophagy [54], which may subsequently induce pro-immunogenic signaling mechanisms. Dying tumor cells have also been shown to release nucleic acids such as RNA that bind to toll-like receptor 3 (TLR3) and activate innate immune cells to secrete type-1 interferons (IFN-1s) [55,56]. IFN-1s have been implicated in the stimulation of both the innate and adaptive anti-tumor immune response and are currently being investigated as a potential target for novel therapies [56].
Enhancing ICD in melanoma patients may lead to a better patient prognosis. Ren et al. identified a group of three differentially expressed genes (GBP2, THBS4, and APOBEC3G) in melanoma patients that are related to ICD and can be utilized to predict patient responses to immunotherapies [57]. Further investigation has shown a positive correlation of tumor-cell ICD with improved patient prognosis [57]. Adjuvant therapies that induce ICD have already been shown to act synergistically with current checkpoint inhibitor therapies by activating DC antigen presentation and T-cell infiltration within the TIME [58]. Hence, focal ICD-inducing therapies may sensitize tumors to immunotherapies and may be effective in combination therapies that improve patient quality of life without adding the systemic adverse effects of systemic chemotherapy.

3. Targets of ICD

3.1. Transcriptional Activators and Epigenetic Targets

To better identify potential targets that can be utilized to promote an anti-tumor immune response, it is important to first understand the mechanisms underlying melanoma cell growth and development. Melanoma stems from the pigment-producing cells of the skin, called melanocytes. One of the transcription factors that plays an important role in melanocyte development, proliferation, and survival is the microphthalmia-associated transcription factor (MITF) [59]. High levels of MITF expression have been shown to favor a non-invasive, proliferative melanoma phenotype, while low levels of MITF expression favor an invasive, metastatic melanoma phenotype [59]. In melanoma patients, the signal transducer and activator of transcription 3 (STAT3), which is known to antagonize MITF expression [60], has been found to play a role in melanoma ICD. One study found that using stattic, a STAT3 inhibitor, in combination with the chemotherapeutic and known ICD-inducer doxorubicin, leads to increased ecto-CRT, increased release of HMGB1, and increased expression of 70-kilodalton heat shock proteins (HSP70) [61], suggesting that STAT3 is a potential target for the induction of melanoma ICD.
Another potential target to increase melanoma tumor immunogenicity is the transcriptional activator SOX10, which also regulates MITF [62,63]. SOX10 plays an important role in neural crest development and inhibits melanoma cell immunogenicity by inducing the expression of interferon regulatory factor 4 (IRF4), a negative regulator of interferon regulatory factor 1 (IRF1) transcription [64]. IRF1 is a tumor suppressor known to promote the activation of IFN-1 genes, another hallmark of ICD [65]. Since SOX10 has been shown to negatively correlate with PD-L1 expression on tumor cells, targeting SOX10 may not only enhance tumor cell ICD, but may also improve responses to anti-PD-1/-PD-L1 therapies by its direct effects on PD-L1 expression [64]. Therapeutics such as histone deacetylase inhibitors may be used to suppress SOX10 expression, thus leading to the induction of PD-L1 expression on melanoma cells and increased responsiveness to anti-PD-L1 therapies [64,66].
In addition to histone deacetylase inhibitors, which have been shown to improve melanoma cell responsiveness to therapy, other epigenetic modulations have also been shown to promote immunogenicity within the TIME. For example, a dual inhibitor of histone and DNA methyltransferases was shown to induce apoptosis in B16-F10 melanoma and subsequently resulted in increased activation of CD4+ and CD8+ T-cells within the TIME and tumor growth delay [67]. The chromatin modifier lysine-specific demethylase 1 (LSD1) is an example of an epigenetic target that has also been shown to inhibit anti-tumor immunity [68]. Sheng et al. showed that B16 melanoma tumors that were deficient in LSD1 had increased secretion of IFN-1s, leading to increased T-cell infiltration within the TIME [68]. Additionally, the LSD1-deficient melanoma cells had increased sensitivity to anti-PD-1 therapy [68], suggesting that epigenetic targets that promote ICD marker expression may be utilized to promote tumor immunogenicity and to improve responsiveness to CI therapy.

3.2. ICD Signaling Pathway Targets

ICD is associated with the accumulation of misfolded and unfolded proteins in the ER, resulting in the induction of ER stress and the UPR. The protein kinase RNA-like ER kinase (PERK) arm of the UPR plays a role in ICD [69,70]. PERK has downstream effects on both pro-apoptotic and pro-survival pathways. It also induces the translation of activating transcription factor 4 (ATF4), which plays a key role in the regulation of autophagy [71]. Conversely, the ablation of PERK signaling within melanoma cells induces paraptosis, a specific type of programmed cell death that is also associated with ER stress, increased secretion of IFN-1, and the migration of inflammatory DCs in the TIME [72].
Other types of cell death have also been associated with ICD and an increased susceptibility to CI therapy [73,74]. Ferroptosis, a form of iron-dependent, non-apoptotic cell death, leads to the accumulation of reactive oxygen species and is emerging as a promising target for adjuvant therapies [75]. Regulators of ferroptosis have shown promise as therapeutic targets. For example, calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2) inhibits ferroptosis [76,77], and its expression has been identified as a potential driver in CI therapy resistance [78]. This suggests that targeting CAMKK2 and its signaling pathways may subsequently enhance ferroptosis and immunogenicity within the TIME.
ICD can also be induced via innate immune system pathways. For example, retinoic acid-inducible gene I (RIG-I) is a cytosolic RNA sensor that, when stimulated, can induce ICD via the interferon-dependent apoptosis of melanoma cells [79,80,81]. RIG-I agonists, such as M8, can be used to activate the RIG-I pathway, subsequently activating the innate immune system by increasing DC expression of the co-stimulatory factors CD80 and CD86, and further enhancing DCs’ ability to process antigens and cross-present to CD8+ T-cells [79,80,81].
TLRs are an example of pattern-recognition receptors that recognize pathogen-associated molecular patterns and DAMPs. They also play a major role in initiating the innate immune response and, as a result, are an attractive target for increasing susceptibility to current CI therapies [82,83,84]. In addition to promoting an innate immune response within the TIME, TLR agonists, such as the TLR-2/3 agonist, L-pampo (LP) [85], and the TLR-7 agonist, Imiquimod [86], have been shown to induce tumor-cell ICD, as evidenced by enhanced ecto-CRT and the increased release of HMGB1 and ATP [85,86]. Both TLR agonists promote a CD8+ T-cell- and IFNγ-driven anti-tumor response and lead to increased CD8+ T-cell infiltration in B16-F10 tumors [86].
The direct targeting of molecules and mechanisms known to play a role in ICD (Table 2) may enhance the efficient induction of an anti-tumor adaptive immune response. However, depending on the immunogenicity of a tumor and its associated mutations, sometimes, multiple mechanisms may be needed to promote pro-immunogenic signaling within the TIME. Additional therapies that induce tumor-cell ICD by targeting alternative mechanisms (Table 3) may be used as adjuvants to enhance the endogenous anti-tumor response and to increase patient responses to current gold standard therapies.

4. Novel Pro-Immunogenic Adjuvant Therapies

4.1. Nanovesicles for Local Targeting of Chemical ICD-Inducers

Some chemotherapeutics, including doxorubicin and paclitaxel, promote anti-tumor immunity by promoting ICD [96]. Doxorubicin has also been shown to increase the expression of PD-L1 by tumor cells [87], which may promote T-cell exhaustion within the TIME [87]. Combination therapies with ICD inducers may enhance the responsiveness to anti-PD-L1 therapy. Celastrol, a member of the quinone methide family, may enhance the immune response via multiple pathways, including the induction of tumor cell autophagy, suppression of PD-L1 expression by melanoma cells, and induction of ICD [88].
Other ICD-inducers under investigation for use in melanoma include chromomycins A5-8, antibiotics that induce tumor cell ICD, autophagy, apoptosis, and ecto-CRT [89]; and polyphenols, which promote the release of calcium from the ER and subsequently disrupt the mitochondrial membrane potential, leading to ICD [90]. Another ICD-inducer under investigation is peptide RT53, which induced ICD and complete tumor regression in a B16-F10 mouse model [91].
However, one of the challenges with chemical inducers of ICD is the potential for severe side effects, including cardiotoxicity and neurotoxicity [97]. Targeted therapies that allow the local delivery of ICD-inducers directly to the tumor site are a major area of research. Nanovesicles are an attractive therapeutic tool since they can be conjugated with targeting ligands and can be modulated to retain their cargo until they reach the tumor site. Acidic pH or near-infrared (NIR) light can then trigger the intra-tumoral release of ICD-inducers from nanovesicles [98], thereby mitigating the systemic, off-target adverse effects. Studies have shown that the nanovesicular delivery of ICD-inducers leads to increased activation of DCs as compared to when ICD-inducers are delivered without nanovesicular targeting [97]. Nanovesicles have the added benefit of allowing the loading and targeted delivery of combinatorial treatments. One group designed a nanovesicular platform that is loaded with multiple ICD-inducers in addition to an indoleamine 2,3-dioxygenase (IDO) inhibitor [99]. This combination therapy takes advantage of another mechanistic target of anti-tumor therapies: metabolism. The metabolic profiles of tumor cells influence their ability to activate the immune response and affect immune cell functions [47,100,101,102,103,104,105]. For example, tumor cells expressing the tryptophan-catabolizing enzyme IDO are known to lead to T-cell dysfunction [36,106,107,108]. IDO, therefore, is an attractive target that may complement ICD’s effects on the adaptive immune response. Certain compounds, such as the tryptanthrin derivative CY-1-4, both inhibit IDO and promote ICD [109] and have been investigated in melanoma models as potential therapeutics.

4.2. Oncolytic Viruses as Cancer Vaccines

Oncolytic viruses are of interest due to their ability to induce and secrete DAMPs and tumor antigens, while reprogramming and immune-activating the stromal cells in the TIME [110]. These viruses infect and lyse cancer cells and have already been used in human patients with promising results. Talimogene laherparepvec (T-VEC), an oncolytic herpes simplex virus HSV-1 that encodes the granulocyte-macrophage colony-stimulating factor, has already been FDA-approved for the treatment of melanoma [111]. Although the mechanism of action has not yet been fully characterized, T-VEC has been shown to promote ICD by increasing ecto-CRT and the release of HMGB1 and ATP [111]. Researchers have also found that the response to T-VEC appears to be inversely correlated with the expression of the stimulator of interferon genes (STING). When melanoma cells are deficient in the STING, there is increased tumor cell death in response to T-VEC which leads to increased CD8+ T-cell recruitment and infiltration to the tumor site [92,93]. Further modifying current oncolytic virus therapies to encode additional fusion proteins, such as the highly fusogenic form of the envelope glycoprotein of gibbon ape leukemia virus (GALV-GP-R-), can further enhance their immunogenic effect by promoting abscopal effects and the memory T-cell response [94]. Targeting ligands can also be used to further enhance the pro-immunogenic effects of cancer vaccines. One such potential target is CD47, a membrane protein expressed by tumor cells that prevents them from being recognized and phagocytosed by macrophages [112,113]. An adenovirus-based tumor vaccine loaded with a CD47-targeting nanobody fused with the IgG2a Fc protein was found to induce anti-tumoral immunity and promote tumor regression and overall long-term survival in mice [114].
Other oncolytic viruses have also been evaluated as promising immunomodulatory tools to create a “hot” pro-immunogenic tumor microenvironment [115,116,117]. Although not all of these act by inducing ICD, they are being investigated as complementary therapies that work synergistically with ICD-inducers. For example, FixVac (BNT111), an intravenously administered liposomal RNA vaccine (Clinical Trial NCT02410733), was shown to enhance anti-tumor immunity when used in combination with ICD-inducers [95].

4.3. Carbon Ion Radiotherapy

Carbon ion radiotherapy (CIRT), also called heavy ion therapy, is an emerging form of radiation therapy that uses carbon ion/nuclei beam radiation to primarily treat solid tumors. One of the advantages of CIRT is its increased precision in targeting the tumor site, thus mitigating off-target adverse effects due to the characteristic energy distribution of particles, known as the “Bragg Peak” [118]. CIRT also has a greater relative biological effectiveness compared to other forms of radiation therapy since the damage caused by the carbon ions is highly focused in the DNA and thus is able to efficiently overwhelm the cellular stress response [118]. As of January 2023, there are currently 14 centers worldwide that provide patients with CIRT [119]. Primarily used in the treatment of solid tumors such as melanoma, CIRT promotes tumor immunogenicity by enhancing the translocation of calreticulin to the plasma membrane, enhancing the release of HMGB1 and ATP, and promoting an IFN1 response, all major hallmarks of ICD [120]. CIRT has currently been investigated in melanoma in combination with anti-PD-1 therapy and was shown to enhance CD4+ and CD8+ tumor infiltration [120]. It has also been shown to decrease populations of immunosuppressive cells, such as myeloid-derived suppressor cells, within the TIME, further supporting a pro-immunogenic effect [121]. In clinical trials, CIRT has also been shown to improve patients’ progression-free survival, suggesting that it is a promising adjuvant treatment to current immunotherapies [122].

4.4. Photodynamic and Photothermal Therapy

Phototherapy relies on the application of a photosensitizer, typically a chemical compound or nanoparticle, that is excited by a light source to generate either local hyperthermia (photothermal therapy) or reactive oxygen species (photodynamic therapy) at a desired target location. These stressors subsequently affect cellular homeostasis and activate cellular stress responses and ICD [123]. Recently, various photosynthetic drugs and nanoparticle drug delivery systems for the treatment of melanoma were reviewed [124]. Known ICD-inducers such as doxorubicin have already been paired with photothermal agents such as indocyanine green as a mechanism to further sensitize tumor cells to ICD induction [125]. Current research is focused on identifying novel photosensitizers that can be administered for phototherapy, often in the form of multi-component nanovesicles or polymeric networks in order to optimize targeting to the tumor site [126,127]. For example, Konda et al. identified two NIR-absorbing ruthenium (II) complexes, ML19B01 and ML19B02, that promote tumor cell ICD upon activation by NIR [128]. Graphene oxide nanosheets are also an example of a photosensitizer, which, upon exposure to NIR light, induce local hyperthermia at the tumor site, leading to an increased expression of heat shock proteins and subsequent ICD [129].
Studies to enhance photothermal therapy effects have also investigated other photosensitizing mechanisms. Optical droplet vaporization is a strategy that was previously used to improve ultrasound imaging, in which perfluorocarbon droplets are superheated and vaporized to a gas phase [130]. This procedure was also found to cause direct cellular damage and activate cellular stress responses and, as a result, is now also applied with photothermal therapy [130].
Photodynamic therapy of B16 melanoma leads to enhanced ecto-CRT and an increased release of DAMPs, including HMGB1 and IFN1 [131]. Phototherapy also leads to additional downstream effects on the anti-tumor immune response, including the enhanced phagocytic activity of monocytes and enhanced cross-presentation to CD8+ T-cells, thus increasing CD8+ T-cell activation and proliferation [131]. The overall goal of phototherapy is to precisely target the tumor site in order to mitigate off-target adverse effects. Additionally, the immunogenic effects seen with phototherapy suggest that this therapy holds promise as a tool that can remodel the TIME and induce systemic, tumor-specific immunity.

4.5. Focused Ultrasound

Therapeutic focused ultrasounds (FUSs) can be used as a non-invasive adjuvant therapy to induce thermal and/or mechanical stress focally at the tumor site. A piezoelectric transducer is used to convert an electrical signal into ultrasonic waves, characterized by periods of compression and periods of rarefaction, measured by the peak positive pressure and peak negative pressure, respectively. High-intensity FUS (HIFU), where the delivered total energy intensity is typically greater than 1000 Watts/cm2, has already been applied in clinical trials for various solid cancers. HIFU primarily acts by inducing thermal coagulative necrosis, which is associated with non-specific tissue destruction and inflammation, and the induction of cellular stress responses [132,133,134,135,136]. FUS has been shown to promote a T-cell-mediated anti-tumor response, characterized by decreased tumor growth and proliferation [137]; increased infiltration of activated, tumor-specific effector CD4+ and CD8+ T-cells; and increased infiltration of inflammatory macrophages [138,139]. In a B16-F10 melanoma mouse model, the combination therapy of FUS with an anti-CD40 agonistic antibody activated DCs by upregulating costimulatory molecules (ex: CD80, CD86) and MHC class II expression [139]. This subsequently induced melanoma-specific T-cell immunity and also suppressed the growth of a secondary, untreated tumor, suggesting that FUS may promote an abscopal effect by inducing systemic anti-tumoral immunity [139].
In addition to promoting immunogenicity at the tumor site, FUS has also been combined with nanobubbles to enhance drug delivery. The nanobubbles contain inert gas, which, upon application of FUS, begin to oscillate, expanding and collapsing, generating a phenomenon called the cavitation effect [140]. The delivery of ultrasound pulses at the tumor site and subsequent oscillations cause increased permeability of the affected cells’ membranes, increasing both the number and the size of membrane pores and facilitating drug delivery and efficacy [141,142]. When given in combination with CI therapy, the adjuvant FUS + nanobubble treatment enhances the CD8+ T-cell anti-tumor response and generates long-term memory, suggesting that it may likewise contribute to an abscopal effect [140]. This parallels the effects of another treatment modality called electrochemotherapy, in which an electrical field is applied to destabilize the cell membrane and improve drug diffusion into cells [143]. FUS is one of few non-invasive adjuvant therapies that can be used to enhance the T-cell-mediated anti-tumor response both by inducing hyperthermia and increasing membrane permeability.

4.6. Sonodynamic Therapy

FUS has also gained interest within the context of sonodynamic therapy, which refers to a combination therapy using FUS with an intra-tumoral injection of a sensitizing agent, such as a nickel ferrite/carbon nanocomposite [144]. This leads to both increased ICD as well as increased membrane permeability for synergistic, pro-immunogenic effects. Sonodynamic therapy (SDT) has been used to target brain metastasis in melanoma and can promote tumor cell damage and apoptosis via the generation of reactive oxygen species and cavitation [145]. When applied in vivo, SDT induced approximately 60% necrosis in a B16-F10 tumor model [144]. When FUS was combined with doxorubicin-releasing microspheres, it was found to extend progression-free survival in a melanoma model, further emphasizing the promising potential of these dual, pro-immunogenic therapies [146].

4.7. Magnetic Hyperthermia

Magnetic hyperthermia is a form of nanovesicle-mediated, localized hyperthermia that induces tumor cell death. Magnetic hyperthermia delivered via Fe3O4 nanoparticles has been shown to induce necrotic ICD and has been used both in preclinical mouse studies and in preliminary clinical trials in patients with advanced metastatic melanoma [147,148]. In mouse melanoma models, magnetic hyperthermia combined with CI therapy led to enhanced tumor regression compared to CI therapy alone. Furthermore, its combination with radiotherapy increased the expression of chemoattractants and the activation of TLR pathways in the TIME, indicating that magnetic hyperthermia can enhance the immune activation in TIME [147,148]. In a preliminary clinical trial of magnetic hyperthermia currently underway at the Sapporo Medical University in Japan (Clinical Trial Research Protocol No. 18-67), of the four patients treated so far, one patient showed complete tumor regression, and one patient showed a partial anti-tumor response [148]. While further studies are still needed, magnetic hyperthermia remains a prospective tool to locally induce tumor ICD and enhance immunogenicity within the TIME.
Table 3. Treatment modalities that promote ICD and their mechanisms of action.
Table 3. Treatment modalities that promote ICD and their mechanisms of action.
ICD Therapy PlatformMechanismReferences
NanovesiclesPlatform that utilizes targeting ligands to deliver cargo to tumor site; mitigate systemic adverse effects; acidic pH or NIR can trigger cargo release; allow co-loading of cargo with other sensitizers/ICD-inducers; leads to increased DC activation[97,99]
Carbon ion radiotherapy (CIRT)Carbon ion (12C6+) beam radiation is directed at the tumor site; enhances CD4+ and CD8+ tumor infiltration; decreases myeloid-derived suppressor cell infiltration of the TIME[120,121]
Photodynamic/photothermal therapyPhotosensitizer is excited by a light source to generate local hyperthermia (photothermal therapy) or ROS (photodynamic therapy) at the tumor site[123]
Focused ultrasound (FUS)Leads to non-specific tissue destruction and inflammation (HIFU) or to mild hyperthermia with increased heat shock protein expression and induction of cellular stress responses (LOFU); combination with nanovesicles and microbubbles leads to increased cell membrane permeability and improved drug delivery[132,133,134,135,136,140]
Sonodynamic therapyCombination therapy of FUS with a sensitizing agent; generates ROS and promotes tumor cell damage, apoptosis, and necrosis[145]
Magnetic hyperthermiaNanovesicle-mediated, localized hyperthermia induces necrotic ICD; increases expression of chemoattractant and TLR pathway markers[147,148]
Nanosecond pulsed electric fieldsElectric pulses enhance membrane permeability and trigger cellular stress responses (autophagy, necrosis, and apoptosis)[149,150]
Plasma-derived oxidantsIncreases presence of ROS/RNS[151,152]

4.8. Nanosecond Pulsed Electric Fields

Nanosecond pulsed electric fields (nsPEFs) are another potential adjuvant therapy that can be applied to ablate melanoma tumors. However, studies have shown a high variation in tumor response, likely due to differences in the electrical field parameters among research groups [149,150,153]. While some studies suggest that nsPEFs increase immunogenicity and the induction of cellular processes such as autophagy [149,150], recent work by Rossi et al. suggests that necrosis may be the primary mechanism of cell death resulting from nsPEFs [153]. Overall, nsPEF treatment appears to enhance membrane permeability and trigger cellular stress responses, including autophagy, necrosis, and apoptosis [149,150]. Additional studies are needed to better identify the effect of nsPEF on melanoma tumor models and to determine the efficacy of nsPEF as a tool to enhance TIME immunogenicity.

4.9. Plasma-Derived Pro-Oxidant Treatment Modalities

Partially ionized gas or plasma has been used for biological and medical applications for several decades. For example, a hot argon plasma coagulator is used to cauterize telangiectasia to stop rectal bleeding from radiation proctitis. In contrast, cold physical plasma generates a multitude of reactive oxygen and nitrogen species (ROS/RNS) that can cause mitochondrial and ER stress and ICD in tumor cells, such as melanoma. When chemotherapeutics such as doxorubicin, epirubicin, and oxaliplatin are combined with cold plasma-derived oxidants, they upregulate organic cationic transporter SLC22A16 that facilitates cytotoxic drug uptake, increases the secretion of ICD-associated DAMPs such as ATP, and enhances the tumoricidal effects of chemotherapeutics [154]. Non-thermal plasma is therefore under investigation as an inducer of ICD and potential adjuvant therapy, primarily due to the presence of ICD stressors, specifically ROS/RNS [151,152].

5. Limitations of Current Therapies

Many of the above therapeutic modalities have been shown to promote the adaptive immune response by enhancing the pro-immunogenic ICD. This is especially important within the TIME, which is typically characterized by immunosuppressive cell phenotypes. For example, a major mechanism underlying T-cell tolerance in the TIME involves immature DCs, which have a low expression of co-stimulatory molecules and can lead to the establishment of T-cell anergy [34]. Anergic or tolerant T-cells are defined by transcriptome reprogramming which leads to an unresponsive state with notably reduced cytokine secretion and proliferation [35,37,155]. Similarly, exhausted T-cells, generated in the TIME due to repeated antigen exposure, also secrete lower amounts of effector cytokines and are unable to induce a robust and long-term anti-tumor response [37,38]. In melanoma, intra-tumoral populations of T-cells often show anergic and exhausted phenotypes, and these populations have been associated with increased melanoma persistence [34,36,38].
While ICD-inducing, ablative therapies have been shown to promote tumor immunogenicity, they are still primarily used as focal therapies for the local ablation of tumors. The true test of their efficacy will be in their ability to reprogram the immunosuppressive features of the TIME and to sustain an immune-activating TIME that will facilitate tumor antigen presentation and the cross-priming of both CD4+ and CD8+ T cells. A sustained anti-tumoral immune response that can achieve control of systemic metastases after the induction of ICD in tumor cells by focal ablative therapies has not been commonly observed [156]. To date, there is also limited knowledge on whether ICD-inducing therapies are able to reverse or prevent T-cell anergy. Thus, further work is still required to determine whether these ICD-inducing therapeutic modalities may adequately establish long-term anti-tumor function.

6. Low-Energy Focused Ultrasound (LOFU) for Immune-Priming of ICD-Inducing Therapies

LOFUs are performed by applying a lower-intensity ultrasound to tissue, with a maximal intensity limit of 800 Watts/cm2. Unlike HIFU, which is already being applied as a form of ablative cancer therapy for certain solid malignancies in the United States, LOFU is non-invasive and primarily induces a thermal and mechanical cellular stress response, activating the pre-apoptotic phase of ICD and increasing the expression of heat shock proteins [157,158]. When applied in a B16 murine melanoma tumor model, LOFU, combined with an ablative radiation therapy, was found to lead to long-term, T-cell-dependent control of melanoma tumor growth [157]. The most promising aspect of LOFU treatment is its potential to serve as a source of immune priming. In a B16 melanoma model, LOFU treatment, compared to no treatment, led to a decreased transcriptional expression of multiple anergy-associated genes, including three E3 ubiquitin ligase genes named GRAIL, Cbl-b, and Itch within CD4+ T-cells isolated from tumor-draining lymph nodes [157]. All these E3 ubiquitin ligases were previously shown to be transcriptionally upregulated when T-cell anergy was induced via sustained calcium-calcineurin signaling [159], thus suggesting that LOFU treatment, upon decreasing their transcription, may promote a reversal of anergy in tumor-draining CD4+ T cells. These findings set LOFU aside from other current ICD-inducing therapy modalities, which have not yet been shown to reverse T-cell tolerance. Although the mechanisms by which LOFU may reverse T-cell anergy have not yet been fully characterized, future work in this area is vital to identify targets to overcome the immunosuppressive TIME and to prevent future tumor growth/recurrence.

7. Conclusions

The current gold standard therapies for melanoma are not sufficient to overcome the poor survival rates seen in advanced-stage melanoma patients. Considering the low response rate, the potential for tumor recurrence following therapy, and the immune-related adverse effects associated with CI therapies, there remains a need for adjuvant therapies to further improve patient survival and quality of life. CD4+ and CD8+ T-cells are considered the primary effectors of the anti-tumor response, but in the setting of melanoma, they often enter an anergic and exhausted state. An attractive approach to overcome the tolerant TIME is to promote an endogenous immune response by introducing DAMPs and activating pro-immunogenic signaling pathways. ICD has therefore been garnering more attention as a vital mechanism to promote the anti-tumor response. Therapies that induce ICD stressors directly at the tumor site have the potential to simultaneously enhance the endogenous anti-tumor immune response while mitigating off-target adverse effects. Of the discussed ICD-inducing treatment modalities, LOFU especially shows promise in its ability to reprogram the TIME and promote long-term, pro-immunogenic anti-tumor effects due to its potential to reverse T-cell anergy. Furthermore, LOFU can be combined as an immune-priming therapy prior to traditional, focal ablative therapies. Future research to better understand how these various therapies induce ICD is needed to continue enhancing tumor immunogenicity and to provide novel targets for future treatment strategies.

Author Contributions

Writing—original draft preparation, review and editing, S.P. and C.G.; Funding acquisition—C.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by NCI grant number 1R01CA226861.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

C.G. is a co-founder of BioConvergent Health.

References

  1. Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer statistics, 2023. CA A Cancer J. Clin. 2023, 73, 17–48. [Google Scholar] [CrossRef]
  2. Davis, L.E.; Shalin, S.C.; Tackett, A.J. Current state of melanoma diagnosis and treatment. Cancer Biol. Ther. 2019, 20, 1366–1379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Chocarro, L.; Bocanegra, A.; Blanco, E.; Fernández-Rubio, L.; Arasanz, H.; Echaide, M.; Garnica, M.; Ramos, P.; Piñeiro-Hermida, S.; Vera, R.; et al. Cutting-Edge: Preclinical and Clinical Development of the First Approved Lag-3 Inhibitor. Cells 2022, 11, 2531. [Google Scholar] [CrossRef] [PubMed]
  4. Seth, R.; Messersmith, H.; Kaur, V.; Kirkwood, J.M.; Kudchadkar, R.; McQuade, J.L.; Provenzano, A.; Swami, U.; Weber, J.; Alluri, K.C.; et al. Systemic Therapy for Melanoma: ASCO Guideline. J. Clin. Oncol. 2020, 38, 3947–3970. [Google Scholar] [CrossRef] [PubMed]
  5. Wolchok, J.D.; Hodi, F.S.; Weber, J.S.; Allison, J.P.; Urba, W.J.; Robert, C.; O′Day, S.J.; Hoos, A.; Humphrey, R.; Berman, D.M.; et al. Development of ipilimumab: A novel immunotherapeutic approach for the treatment of advanced melanoma. Ann. N. Y. Acad. Sci. 2013, 1291, 1–13. [Google Scholar] [CrossRef] [Green Version]
  6. Barone, A.; Hazarika, M.; Theoret, M.R.; Mishra-Kalyani, P.; Chen, H.; He, K.; Sridhara, R.; Subramaniam, S.; Pfuma, E.; Wang, Y.; et al. FDA Approval Summary: Pembrolizumab for the Treatment of Patients with Unresectable or Metastatic Melanoma. Clin. Cancer Res. 2017, 23, 5661–5665. [Google Scholar] [CrossRef] [Green Version]
  7. Beaver, J.A.; Theoret, M.R.; Mushti, S.; He, K.; Libeg, M.; Goldberg, K.; Sridhara, R.; McKee, A.E.; Keegan, P.; Pazdur, R. FDA Approval of Nivolumab for the First-Line Treatment of Patients with BRAF(V600) Wild-Type Unresectable or Metastatic Melanoma. Clin. Cancer Res. 2017, 23, 3479–3483. [Google Scholar] [CrossRef] [Green Version]
  8. Sun, J.; Zager, J.S.; Eroglu, Z. Encorafenib/binimetinib for the treatment of BRAF-mutant advanced, unresectable, or metastatic melanoma: Design, development, and potential place in therapy. OncoTargets Ther. 2018, 11, 9081–9089. [Google Scholar] [CrossRef] [Green Version]
  9. Ascierto, P.A.; McArthur, G.A.; Dréno, B.; Atkinson, V.; Liszkay, G.; Di Giacomo, A.M.; Mandalà, M.; Demidov, L.; Stroyakovskiy, D.; Thomas, L.; et al. Cobimetinib combined with vemurafenib in advanced BRAF(V600)-mutant melanoma (coBRIM): Updated efficacy results from a randomised, double-blind, phase 3 trial. Lancet Oncol. 2016, 17, 1248–1260. [Google Scholar] [CrossRef]
  10. Webster, R.M.; Mentzer, S.E. The malignant melanoma landscape. Nat. Rev. Drug Discov. 2014, 13, 491–492. [Google Scholar] [CrossRef]
  11. Flaherty, K.T.; Robert, C.; Hersey, P.; Nathan, P.; Garbe, C.; Milhem, M.; Demidov, L.V.; Hassel, J.C.; Rutkowski, P.; Mohr, P.; et al. Improved survival with MEK inhibition in BRAF-mutated melanoma. N. Engl. J. Med. 2012, 367, 107–114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Eggermont, A.M.; Kirkwood, J.M. Re-evaluating the role of dacarbazine in metastatic melanoma: What have we learned in 30 years? Eur. J. Cancer 2004, 40, 1825–1836. [Google Scholar] [CrossRef] [PubMed]
  13. Atkins, M.B.; Lotze, M.T.; Dutcher, J.P.; Fisher, R.I.; Weiss, G.; Margolin, K.; Abrams, J.; Sznol, M.; Parkinson, D.; Hawkins, M.; et al. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: Analysis of 270 patients treated between 1985 and 1993. J. Clin. Oncol. 1999, 17, 2105–2116. [Google Scholar] [CrossRef]
  14. Eggermont, A.M.; Suciu, S.; Santinami, M.; Testori, A.; Kruit, W.H.; Marsden, J.; Punt, C.J.; Salès, F.; Gore, M.; MacKie, R.; et al. Adjuvant therapy with pegylated interferon alfa-2b versus observation alone in resected stage III melanoma: Final results of EORTC 18991, a randomised phase III trial. Lancet 2008, 372, 117–126. [Google Scholar] [CrossRef] [Green Version]
  15. Luke, J.J.; Flaherty, K.T.; Ribas, A.; Long, G.V. Targeted agents and immunotherapies: Optimizing outcomes in melanoma. Nat. Rev. Clin. Oncol. 2017, 14, 463–482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Sullivan, R.J.; Atkins, M.B.; Kirkwood, J.M.; Agarwala, S.S.; Clark, J.I.; Ernstoff, M.S.; Fecher, L.; Gajewski, T.F.; Gastman, B.; Lawson, D.H.; et al. An update on the Society for Immunotherapy of Cancer consensus statement on tumor immunotherapy for the treatment of cutaneous melanoma: Version 2.0. J. Immunother. Cancer 2018, 6, 44. [Google Scholar] [CrossRef] [Green Version]
  17. Seidel, J.A.; Otsuka, A.; Kabashima, K. Anti-PD-1 and anti-CTLA-4 therapies in cancer: Mechanisms of action, efficacy, and limitations. Front. Oncol. 2018, 8, 86. [Google Scholar] [CrossRef] [PubMed]
  18. Arlauckas, S.P.; Garris, C.S.; Kohler, R.H.; Kitaoka, M.; Cuccarese, M.F.; Yang, K.S.; Miller, M.A.; Carlson, J.C.; Freeman, G.J.; Anthony, R.M.; et al. In vivo imaging reveals a tumor-associated macrophage–mediated resistance pathway in anti–PD-1 therapy. Sci. Transl. Med. 2017, 9, eaal3604. [Google Scholar] [CrossRef] [Green Version]
  19. Chen, G.; Huang, A.C.; Zhang, W.; Zhang, G.; Wu, M.; Xu, W.; Yu, Z.; Yang, J.; Wang, B.; Sun, H.; et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 2018, 560, 382–386. [Google Scholar] [CrossRef]
  20. Tang, H.; Liang, Y.; Anders, R.A.; Taube, J.M.; Qiu, X.; Mulgaonkar, A.; Liu, X.; Harrington, S.M.; Guo, J.; Xin, Y.; et al. PD-L1 on host cells is essential for PD-L1 blockade–mediated tumor regression. J. Clin. Investig. 2018, 128, 580–588. [Google Scholar] [CrossRef] [Green Version]
  21. Lin, H.; Wei, S.; Hurt, E.M.; Green, M.D.; Zhao, L.; Vatan, L.; Szeliga, W.; Herbst, R.; Harms, P.W.; Fecher, L.A.; et al. Host expression of PD-L1 determines efficacy of PD-L1 pathway blockade–mediated tumor regression. J. Clin. Investig. 2018, 128, 805–815. [Google Scholar] [CrossRef] [Green Version]
  22. Tarhini, A.A.; Lee, S.J.; Hodi, F.S.; Rao, U.N.; Cohen, G.I.; Hamid, O.; Hutchins, L.F.; Sosman, J.A.; Kluger, H.M.; Eroglu, Z.; et al. Phase III study of adjuvant ipilimumab (3 or 10 mg/kg) versus high-dose interferon alfa-2b for resected high-risk melanoma: North American Intergroup E1609. J. Clin. Oncol. 2020, 38, 567–575. [Google Scholar] [CrossRef]
  23. Almutairi, A.R.; McBride, A.; Slack, M.; Erstad, B.L.; Abraham, I. Potential immune-related adverse events associated with monotherapy and combination therapy of ipilimumab, nivolumab, and pembrolizumab for advanced melanoma: A systematic review and meta-analysis. Front. Oncol. 2020, 10, 91. [Google Scholar] [CrossRef] [PubMed]
  24. Eggermont, A.M.; Kicinski, M.; Blank, C.U.; Mandala, M.; Long, G.V.; Atkinson, V.; Dalle, S.; Haydon, A.; Khattak, A.; Carlino, M.S.; et al. Association between immune-related adverse events and recurrence-free survival among patients with stage III melanoma randomized to receive pembrolizumab or placebo: A secondary analysis of a randomized clinical trial. JAMA Oncol. 2020, 6, 519–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Davis, A.A.; Patel, V.G. The role of PD-L1 expression as a predictive biomarker: An analysis of all US Food and Drug Administration (FDA) approvals of immune checkpoint inhibitors. J. Immunother. Cancer 2019, 7, 278. [Google Scholar] [CrossRef] [PubMed]
  26. Wang, Y.; Tong, Z.; Zhang, W.; Zhang, W.; Buzdin, A.; Mu, X.; Yan, Q.; Zhao, X.; Chang, H.-H.; Duhon, M.; et al. FDA-approved and emerging next generation predictive biomarkers for immune checkpoint inhibitors in cancer patients. Front. Oncol. 2021, 11, 683419. [Google Scholar] [CrossRef] [PubMed]
  27. Marzagalli, M.; Ebelt, N.D.; Manuel, E.R. Unraveling the crosstalk between melanoma and immune cells in the tumor microenvironment. Semin. Cancer Biol. 2019, 59, 236–250. [Google Scholar] [CrossRef]
  28. Sallusto, F.; Lanzavecchia, A. Understanding dendritic cell and T-lymphocyte traffic through the analysis of chemokine receptor expression. Immunol. Rev. 2000, 177, 134–140. [Google Scholar] [CrossRef]
  29. Hussein, M.R. Dendritic cells and melanoma tumorigenesis: An insight. Cancer Biol. Ther. 2005, 4, 501–505. [Google Scholar] [CrossRef] [Green Version]
  30. Bennaceur, K.; Chapman, J.; Brikci-Nigassa, L.; Sanhadji, K.; Touraine, J.-l.; Portoukalian, J. Dendritic cells dysfunction in tumour environment. Cancer Lett. 2008, 272, 186–196. [Google Scholar] [CrossRef]
  31. Tucci, M.; Stucci, S.; Passarelli, A.; Giudice, G.; Dammacco, F.; Silvestris, F. The immune escape in melanoma: Role of the impaired dendritic cell function. Expert Rev. Clin. Immunol. 2014, 10, 1395–1404. [Google Scholar] [CrossRef] [PubMed]
  32. Monti, M.; Consoli, F.; Vescovi, R.; Bugatti, M.; Vermi, W. Human plasmacytoid dendritic cells and cutaneous melanoma. Cells 2020, 9, 417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Miloud, T.; Hämmerling, G.J.; Garbi, N. Review of murine dendritic cells: Types, location, and development. Dendritic Cell Protoc. 2010, 595, 21–42. [Google Scholar] [CrossRef]
  34. Passarelli, A.; Mannavola, F.; Stucci, L.S.; Tucci, M.; Silvestris, F. Immune system and melanoma biology: A balance between immunosurveillance and immune escape. Oncotarget 2017, 8, 106132–106142. [Google Scholar] [CrossRef] [PubMed]
  35. Schwartz, R.H. T cell anergy. Annu. Rev. Immunol. 2003, 21, 305–334. [Google Scholar] [CrossRef] [PubMed]
  36. Gajewski, T.F. Failure at the effector phase: Immune barriers at the level of the melanoma tumor microenvironment. Clin. Cancer Res. 2007, 13, 5256–5261. [Google Scholar] [CrossRef] [Green Version]
  37. Valdor, R.; Macian, F. Induction and stability of the anergic phenotype in T cells. Semin. Immunol. 2013, 25, 313–320. [Google Scholar] [CrossRef] [Green Version]
  38. Oliveira, G.; Stromhaug, K.; Klaeger, S.; Kula, T.; Frederick, D.T.; Le, P.M.; Forman, J.; Huang, T.; Li, S.; Zhang, W.; et al. Phenotype, specificity and avidity of antitumour CD8+ T cells in melanoma. Nature 2021, 596, 119–125. [Google Scholar] [CrossRef]
  39. Fucikova, J.; Kepp, O.; Kasikova, L.; Petroni, G.; Yamazaki, T.; Liu, P.; Zhao, L.; Spisek, R.; Kroemer, G.; Galluzzi, L. Detection of immunogenic cell death and its relevance for cancer therapy. Cell Death Dis. 2020, 11, 1013. [Google Scholar] [CrossRef]
  40. Galluzzi, L.; Vitale, I.; Warren, S.; Adjemian, S.; Agostinis, P.; Martinez, A.B.; Chan, T.A.; Coukos, G.; Demaria, S.; Deutsch, E.; et al. Consensus guidelines for the definition, detection and interpretation of immunogenic cell death. J. Immunother. Cancer 2020, 8, e000337. [Google Scholar] [CrossRef] [Green Version]
  41. Giglio, P.; Gagliardi, M.; Bernardini, R.; Mattei, M.; Cotella, D.; Santoro, C.; Piacentini, M.; Corazzari, M. Ecto-Calreticulin is essential for an efficient immunogenic cell death stimulation in mouse melanoma. Genes Immun. 2019, 20, 509–513. [Google Scholar] [CrossRef]
  42. Venkateswaran, K.; Verma, A.; Bhatt, A.N.; Shrivastava, A.; Manda, K.; Raj, H.G.; Prasad, A.; Len, C.; Parmar, V.S.; Dwarakanath, B.S. Emerging roles of calreticulin in cancer: Implications for therapy. Curr. Protein Pept. Sci. 2018, 19, 344–357. [Google Scholar] [CrossRef]
  43. Sen Santara, S.; Lee, D.-J.; Crespo, Â.; Hu, J.J.; Walker, C.; Ma, X.; Zhang, Y.; Chowdhury, S.; Meza-Sosa, K.F.; Lewandrowski, M.; et al. The NK cell receptor NKp46 recognizes ecto-calreticulin on ER-stressed cells. Nature 2023, 616, 348–356. [Google Scholar] [CrossRef]
  44. Bronietzki, A.W.; Schuster, M.; Schmitz, I. Autophagy in T-cell development, activation and differentiation. Immunol. Cell Biol. 2015, 93, 25–34. [Google Scholar] [CrossRef] [PubMed]
  45. Dowling, S.D.; Macian, F. Autophagy and T cell metabolism. Cancer Lett. 2018, 419, 20–26. [Google Scholar] [CrossRef]
  46. DeVorkin, L.; Pavey, N.; Carleton, G.; Comber, A.; Ho, C.; Lim, J.; McNamara, E.; Huang, H.; Kim, P.; Zacharias, L.G.; et al. Autophagy regulation of metabolism is required for CD8+ T cell anti-tumor immunity. Cell Rep. 2019, 27, 502–513.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Maes, H.; Rubio, N.; Garg, A.D.; Agostinis, P. Autophagy: Shaping the tumor microenvironment and therapeutic response. Trends Mol. Med. 2013, 19, 428–446. [Google Scholar] [CrossRef]
  48. Messer, J.S. The cellular autophagy/apoptosis checkpoint during inflammation. Cell. Mol. Life Sci. 2017, 74, 1281–1296. [Google Scholar] [CrossRef]
  49. Li, S.; Song, Y.; Quach, C.; Nemecio, D.; Liang, C. Revisiting the role of autophagy in melanoma. Autophagy 2019, 15, 1843–1844. [Google Scholar] [CrossRef] [PubMed]
  50. Prieto, K.; Lozano, M.P.; Urueña, C.; Alméciga-Díaz, C.J.; Fiorentino, S.; Barreto, A. The delay in cell death caused by the induction of autophagy by P2Et extract is essential for the generation of immunogenic signals in melanoma cells. Apoptosis 2020, 25, 875–888. [Google Scholar] [CrossRef]
  51. Martins, I.; Michaud, M.; Sukkurwala, A.Q.; Adjemian, S.; Ma, Y.; Shen, S.; Kepp, O.; Menger, L.; Vacchelli, E.; Galluzzi, L.; et al. Premortem autophagy determines the immunogenicity of chemotherapy-induced cancer cell death. Autophagy 2012, 8, 413–415. [Google Scholar] [CrossRef] [Green Version]
  52. Yuan, J.; Yuan, X.; Wu, K.; Gao, J.; Li, L. A Local and Low-Dose Chemotherapy/Autophagy-Enhancing Regimen Treatment Markedly Inhibited the Growth of Established Solid Tumors through a Systemic Antitumor Immune Response. Front. Oncol. 2021, 11, 658254. [Google Scholar] [CrossRef]
  53. Chen, R.; Kang, R.; Tang, D. The mechanism of HMGB1 secretion and release. Exp. Mol. Med. 2022, 54, 91–102. [Google Scholar] [CrossRef] [PubMed]
  54. Xu, T.; Jiang, L.; Wang, Z. The progression of HMGB1-induced autophagy in cancer biology. OncoTargets Ther. 2019, 12, 365–377. [Google Scholar] [CrossRef] [Green Version]
  55. Vacchelli, E.; Sistigu, A.; Yamazaki, T.; Vitale, I.; Zitvogel, L.; Kroemer, G. Autocrine signaling of type 1 interferons in successful anticancer chemotherapy. Oncoimmunology 2015, 4, e988042. [Google Scholar] [PubMed]
  56. Lamberti, M.J.; Mentucci, F.M.; Roselli, E.; Araya, P.; Rivarola, V.A.; Rumie Vittar, N.B.; Maccioni, M. Photodynamic modulation of type 1 interferon pathway on melanoma cells promotes dendritic cell activation. Front. Immunol. 2019, 10, 2614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Ren, J.; Yang, J.; Na, S.; Wang, Y.; Zhang, L.; Wang, J.; Liu, J. Comprehensive characterisation of immunogenic cell death in melanoma revealing the association with prognosis and tumor immune microenvironment. Front. Immunol. 2022, 13, 998653. [Google Scholar] [CrossRef]
  58. Pfirschke, C.; Engblom, C.; Rickelt, S.; Cortez-Retamozo, V.; Garris, C.; Pucci, F.; Yamazaki, T.; Poirier-Colame, V.; Newton, A.; Redouane, Y.; et al. Immunogenic chemotherapy sensitizes tumors to checkpoint blockade therapy. Immunity 2016, 44, 343–354. [Google Scholar] [CrossRef] [Green Version]
  59. Gelmi, M.C.; Houtzagers, L.E.; Strub, T.; Krossa, I.; Jager, M.J. MITF in normal melanocytes, cutaneous and uveal melanoma: A delicate balance. Int. J. Mol. Sci. 2022, 23, 6001. [Google Scholar] [CrossRef]
  60. Swoboda, A.; Soukup, R.; Eckel, O.; Kinslechner, K.; Wingelhofer, B.; Schörghofer, D.; Sternberg, C.; Pham, H.T.; Vallianou, M.; Horvath, J.; et al. STAT3 promotes melanoma metastasis by CEBP-induced repression of the MITF pathway. Oncogene 2021, 40, 1091–1105. [Google Scholar] [CrossRef]
  61. Jafari, S.; Lavasanifar, A.; Hejazi, M.S.; Maleki-Dizaji, N.; Mesgari, M.; Molavi, O. STAT3 inhibitory stattic enhances immunogenic cell death induced by chemotherapy in cancer cells. DARU J. Pharm. Sci. 2020, 28, 159–169. [Google Scholar] [CrossRef]
  62. Coe, E.A.; Tan, J.Y.; Shapiro, M.; Louphrasitthiphol, P.; Bassett, A.R.; Marques, A.C.; Goding, C.R.; Vance, K.W. The MITF-SOX10 regulated long non-coding RNA DIRC3 is a melanoma tumour suppressor. PLoS Genet. 2019, 15, e1008501. [Google Scholar] [CrossRef] [Green Version]
  63. Beleaua, M.-A.; Jung, I.; Braicu, C.; Milutin, D.; Gurzu, S. SOX11, SOX10 and MITF gene interaction: A possible diagnostic tool in malignant melanoma. Life 2021, 11, 281. [Google Scholar] [CrossRef] [PubMed]
  64. Yokoyama, S.; Takahashi, A.; Kikuchi, R.; Nishibu, S.; Lo, J.A.; Hejna, M.; Moon, W.M.; Kato, S.; Zhou, Y.; Hodi, F.S.; et al. SOX10 Regulates Melanoma Immunogenicity through an IRF4–IRF1 AxisSOX10 Regulates Immunogenicity in Melanoma. Cancer Res. 2021, 81, 6131–6141. [Google Scholar] [CrossRef] [PubMed]
  65. Feng, H.; Zhang, Y.-B.; Gui, J.-F.; Lemon, S.M.; Yamane, D. Interferon regulatory factor 1 (IRF1) and anti-pathogen innate immune responses. PLoS Pathog. 2021, 17, e1009220. [Google Scholar] [CrossRef] [PubMed]
  66. Yokoyama, S.; Feige, E.; Poling, L.L.; Levy, C.; Widlund, H.R.; Khaled, M.; Kung, A.L.; Fisher, D.E. Pharmacologic suppression of MITF expression via HDAC inhibitors in the melanocyte lineage. Pigment Cell Melanoma Res. 2008, 21, 457–463. [Google Scholar] [CrossRef] [Green Version]
  67. De Beck, L.; Awad, R.M.; Basso, V.; Casares, N.; De Ridder, K.; De Vlaeminck, Y.; Gnata, A.; Goyvaerts, C.; Lecocq, Q.; José-Enériz, S.; et al. Inhibiting histone and DNA methylation improves cancer vaccination in an experimental model of melanoma. Front. Immunol. 2022, 13, 799636. [Google Scholar] [CrossRef]
  68. Sheng, W.; LaFleur, M.W.; Nguyen, T.H.; Chen, S.; Chakravarthy, A.; Conway, J.R.; Li, Y.; Chen, H.; Yang, H.; Hsu, P.-H.; et al. LSD1 ablation stimulates anti-tumor immunity and enables checkpoint blockade. Cell 2018, 174, 549–563. [Google Scholar] [CrossRef] [Green Version]
  69. Asadzadeh, Z.; Safarzadeh, E.; Safaei, S.; Baradaran, A.; Mohammadi, A.; Hajiasgharzadeh, K.; Derakhshani, A.; Argentiero, A.; Silvestris, N.; Baradaran, B. Current approaches for combination therapy of cancer: The role of immunogenic cell death. Cancers 2020, 12, 1047. [Google Scholar] [CrossRef] [Green Version]
  70. Rufo, N.; Garg, A.D.; Agostinis, P. The unfolded protein response in immunogenic cell death and cancer immunotherapy. Trends Cancer 2017, 3, 643–658. [Google Scholar] [CrossRef]
  71. Zheng, W.; Xie, W.; Yin, D.; Luo, R.; Liu, M.; Guo, F. ATG5 and ATG7 induced autophagy interplays with UPR via PERK signaling. Cell Commun. Signal. 2019, 17, 42. [Google Scholar] [CrossRef] [Green Version]
  72. Mandula, J.K.; Chang, S.; Mohamed, E.; Jimenez, R.; Sierra-Mondragon, R.A.; Chang, D.C.; Obermayer, A.N.; Moran-Segura, C.M.; Das, S.; Vazquez-Martinez, J.A.; et al. Ablation of the endoplasmic reticulum stress kinase PERK induces paraptosis and type I interferon to promote anti-tumor T cell responses. Cancer Cell 2022, 40, 1145–1160.e9. [Google Scholar] [CrossRef] [PubMed]
  73. Luo, M.; Wu, L.; Zhang, K.; Wang, H.; Zhang, T.; Gutierrez, L.; O’Connell, D.; Zhang, P.; Li, Y.; Gao, T.; et al. miR-137 regulates ferroptosis by targeting glutamine transporter SLC1A5 in melanoma. Cell Death Differ. 2018, 25, 1457–1472. [Google Scholar] [CrossRef] [Green Version]
  74. Wang, W.; Green, M.; Choi, J.E.; Gijón, M.; Kennedy, P.D.; Johnson, J.K.; Liao, P.; Lang, X.; Kryczek, I.; Sell, A.; et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 2019, 569, 270–274. [Google Scholar] [CrossRef]
  75. Li, J.; Cao, F.; Yin, H.-l.; Huang, Z.-j.; Lin, Z.-t.; Mao, N.; Sun, B.; Wang, G. Ferroptosis: Past, present and future. Cell Death Dis. 2020, 11, 88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Dai, X.; Meng, J.; Deng, S.; Zhang, L.; Wan, C.; Lu, L.; Huang, J.; Hu, Y.; Zhang, Z.; Li, Y.; et al. Targeting CAMKII to reprogram tumor-associated macrophages and inhibit tumor cells for cancer immunotherapy with an injectable hybrid peptide hydrogel. Theranostics 2020, 10, 3049–3063. [Google Scholar] [CrossRef]
  77. Wang, S.; Yi, X.; Wu, Z.; Guo, S.; Dai, W.; Wang, H.; Shi, Q.; Zeng, K.; Guo, W.; Li, C. CAMKK2 defines ferroptosis sensitivity of melanoma cells by regulating AMPK–NRF2 pathway. J. Investig. Dermatol. 2022, 142, 189–200. [Google Scholar] [CrossRef]
  78. Tomaszewski, W.H.; Waibl-Polania, J.; Chakraborty, M.; Perera, J.; Ratiu, J.; Miggelbrink, A.; McDonnell, D.P.; Khasraw, M.; Ashley, D.M.; Fecci, P.E.; et al. Neuronal CaMKK2 promotes immunosuppression and checkpoint blockade resistance in glioblastoma. Nat. Commun. 2022, 13, 6483. [Google Scholar] [CrossRef] [PubMed]
  79. Castiello, L.; Zevini, A.; Vulpis, E.; Muscolini, M.; Ferrari, M.; Palermo, E.; Peruzzi, G.; Krapp, C.; Jakobsen, M.; Olagnier, D.; et al. An optimized retinoic acid-inducible gene I agonist M8 induces immunogenic cell death markers in human cancer cells and dendritic cell activation. Cancer Immunol. Immunother. 2019, 68, 1479–1492. [Google Scholar] [CrossRef]
  80. Jiang, X.; Muthusamy, V.; Fedorova, O.; Kong, Y.; Kim, D.J.; Bosenberg, M.; Pyle, A.M.; Iwasaki, A. Intratumoral delivery of RIG-I agonist SLR14 induces robust antitumor responses. J. Exp. Med. 2019, 216, 2854–2868. [Google Scholar] [CrossRef]
  81. Lambing, S.; Tan, Y.P.; Vasileiadou, P.; Holdenrieder, S.; Müller, P.; Hagen, C.; Garbe, S.; Behrendt, R.; Schlee, M.; van den Boorn, J.G.; et al. RIG-I immunotherapy overcomes radioresistance in p53-positive malignant melanoma. J. Mol. Cell Biol. 2023, 15, mjad001. [Google Scholar] [CrossRef] [PubMed]
  82. Rolfo, C.; Giovannetti, E.; Martinez, P.; McCue, S.; Naing, A. Applications and clinical trial landscape using Toll-like receptor agonists to reduce the toll of cancer. NPJ Precis. Oncol. 2023, 7, 26. [Google Scholar] [CrossRef]
  83. Bourquin, C.; Pommier, A.; Hotz, C. Harnessing the immune system to fight cancer with Toll-like receptor and RIG-I-like receptor agonists. Pharmacol. Res. 2020, 154, 104192. [Google Scholar] [CrossRef] [Green Version]
  84. Ribas, A.; Medina, T.; Kirkwood, J.M.; Zakharia, Y.; Gonzalez, R.; Davar, D.; Chmielowski, B.; Campbell, K.M.; Bao, R.; Kelley, H.; et al. Overcoming PD-1 blockade resistance with CpG-A toll-like receptor 9 agonist vidutolimod in patients with metastatic melanoma. Cancer Discov. 2021, 11, 2998–3007. [Google Scholar] [CrossRef] [PubMed]
  85. Lee, W.S.; Kim, D.S.; Kim, J.H.; Heo, Y.; Yang, H.; Go, E.-J.; Kim, J.H.; Lee, S.J.; Ahn, B.C.; Yum, J.S.; et al. Intratumoral immunotherapy using a TLR2/3 agonist, L-pampo, induces robust antitumor immune responses and enhances immune checkpoint blockade. J. Immunother. Cancer 2022, 10, e004799. [Google Scholar] [CrossRef] [PubMed]
  86. Huang, S.-W.; Wang, S.-T.; Chang, S.-H.; Chuang, K.-C.; Wang, H.-Y.; Kao, J.-K.; Liang, S.-M.; Wu, C.-Y.; Kao, S.-H.; Chen, Y.-J.; et al. Imiquimod exerts antitumor effects by inducing immunogenic cell death and is enhanced by the glycolytic inhibitor 2-deoxyglucose. J. Investig. Dermatol. 2020, 140, 1771–1783.e6. [Google Scholar] [CrossRef]
  87. Hu, M.; Zhang, J.; Kong, L.; Yu, Y.; Hu, Q.; Yang, T.; Wang, Y.; Tu, K.; Qiao, Q.; Qin, X.; et al. Immunogenic hybrid nanovesicles of liposomes and tumor-derived nanovesicles for cancer immunochemotherapy. ACS Nano 2021, 15, 3123–3138. [Google Scholar] [CrossRef]
  88. Qiu, N.; Liu, Y.; Liu, Q.; Chen, Y.; Shen, L.; Hu, M.; Zhou, X.; Shen, Y.; Gao, J.; Huang, L. Celastrol nanoemulsion induces immunogenicity and downregulates PD-L1 to boost abscopal effect in melanoma therapy. Biomaterials 2021, 269, 120604. [Google Scholar] [CrossRef]
  89. Florêncio, K.G.D.; Edson, E.A.; da Silva Fernandes, K.S.; Luiz, J.P.M.; Pinto, F.d.C.L.; Pessoa, O.D.L.; de Queiroz Cunha, F.; Machado-Neto, J.A.; Wilke, D.V. Chromomycin A5 induces bona fide immunogenic cell death in melanoma. Front. Immunol. 2022, 13, 941757. [Google Scholar] [CrossRef]
  90. Prieto, K.; Cao, Y.; Mohamed, E.; Trillo-Tinoco, J.; Sierra, R.A.; Urueña, C.; Sandoval, T.A.; Fiorentino, S.; Rodriguez, P.C.; Barreto, A. Polyphenol-rich extract induces apoptosis with immunogenic markers in melanoma cells through the ER stress-associated kinase PERK. Cell Death Discov. 2019, 5, 134. [Google Scholar] [CrossRef] [Green Version]
  91. Pasquereau-Kotula, E.; Habault, J.; Kroemer, G.; Poyet, J.-L. The anticancer peptide RT53 induces immunogenic cell death. PLoS ONE 2018, 13, e0201220. [Google Scholar] [CrossRef]
  92. Bommareddy, P.K.; Zloza, A.; Rabkin, S.D.; Kaufman, H.L. Oncolytic virus immunotherapy induces immunogenic cell death and overcomes STING deficiency in melanoma. Oncoimmunology 2019, 8, 1591875. [Google Scholar] [CrossRef]
  93. Ferrucci, P.F.; Pala, L.; Conforti, F.; Cocorocchio, E. Talimogene laherparepvec (T-VEC): An intralesional cancer immunotherapy for advanced melanoma. Cancers 2021, 13, 1383. [Google Scholar] [CrossRef]
  94. Thomas, S.; Kuncheria, L.; Roulstone, V.; Kyula, J.N.; Mansfield, D.; Bommareddy, P.K.; Smith, H.; Kaufman, H.L.; Harrington, K.J.; Coffin, R.S. Development of a new fusion-enhanced oncolytic immunotherapy platform based on herpes simplex virus type 1. J. Immunother. Cancer 2019, 7, 214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Sahin, U.; Oehm, P.; Derhovanessian, E.; Jabulowsky, R.A.; Vormehr, M.; Gold, M.; Maurus, D.; Schwarck-Kokarakis, D.; Kuhn, A.N.; Omokoko, T.; et al. An RNA vaccine drives immunity in checkpoint-inhibitor-treated melanoma. Nature 2020, 585, 107–112. [Google Scholar] [CrossRef] [PubMed]
  96. Tu, K.; Deng, H.; Kong, L.; Wang, Y.; Yang, T.; Hu, Q.; Hu, M.; Yang, C.; Zhang, Z. Reshaping tumor immune microenvironment through acidity-responsive nanoparticles featured with CRISPR/Cas9-mediated programmed death-ligand 1 attenuation and chemotherapeutics-induced immunogenic cell death. ACS Appl. Mater. Interfaces 2020, 12, 16018–16030. [Google Scholar] [CrossRef]
  97. Liu, M.; Hao, L.; Zhao, D.; Li, J.; Lin, Y. Self-Assembled Immunostimulatory Tetrahedral Framework Nucleic Acid Vehicles for Tumor Chemo-immunotherapy. ACS Appl. Mater. Interfaces 2022, 14, 38506–38514. [Google Scholar] [CrossRef]
  98. Li, Z.; Chu, Z.; Yang, J.; Qian, H.; Xu, J.; Chen, B.; Tian, T.; Chen, H.; Xu, Y.; Wang, F. Immunogenic Cell Death Augmented by Manganese Zinc Sulfide Nanoparticles for Metastatic Melanoma Immunotherapy. ACS Nano 2022, 16, 15471–15483. [Google Scholar] [CrossRef] [PubMed]
  99. Wang, Z.; Little, N.; Chen, J.; Lambesis, K.T.; Le, K.T.; Han, W.; Scott, A.J.; Lu, J. Immunogenic camptothesome nanovesicles comprising sphingomyelin-derived camptothecin bilayers for safe and synergistic cancer immunochemotherapy. Nat. Nanotechnol. 2021, 16, 1130–1140. [Google Scholar] [CrossRef]
  100. Dupont, N.; Jiang, S.; Pilli, M.; Ornatowski, W.; Bhattacharya, D.; Deretic, V. Autophagy-based unconventional secretory pathway for extracellular delivery of IL-1β. EMBO J. 2011, 30, 4701–4711. [Google Scholar] [CrossRef] [Green Version]
  101. Michaud, M.; Martins, I.; Sukkurwala, A.Q.; Adjemian, S.; Ma, Y.; Pellegatti, P.; Shen, S.; Kepp, O.; Scoazec, M.; Mignot, G.; et al. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science 2011, 334, 1573–1577. [Google Scholar] [CrossRef]
  102. Garg, A.D.; Galluzzi, L.; Apetoh, L.; Baert, T.; Birge, R.B.; Bravo-San Pedro, J.M.; Breckpot, K.; Brough, D.; Chaurio, R.; Cirone, M.; et al. Molecular and translational classifications of DAMPs in immunogenic cell death. Front. Immunol. 2015, 6, 588. [Google Scholar] [CrossRef] [Green Version]
  103. Huang, C.; Radi, R.H.; Arbiser, J.L. Mitochondrial metabolism in melanoma. Cells 2021, 10, 3197. [Google Scholar] [CrossRef]
  104. Harel, M.; Ortenberg, R.; Varanasi, S.K.; Mangalhara, K.C.; Mardamshina, M.; Markovits, E.; Baruch, E.N.; Tripple, V.; Arama-Chayoth, M.; Greenberg, E.; et al. Proteomics of melanoma response to immunotherapy reveals mitochondrial dependence. Cell 2019, 179, 236–250.e218. [Google Scholar] [CrossRef]
  105. Hargadon, K.M. Strategies to improve the efficacy of dendritic cell-based immunotherapy for melanoma. Front. Immunol. 2017, 8, 1594. [Google Scholar] [CrossRef] [Green Version]
  106. Fallarino, F.; Grohmann, U.; Vacca, C.; Bianchi, R.; Orabona, C.; Spreca, A.; Fioretti, M.; Puccetti, P. T cell apoptosis by tryptophan catabolism. Cell Death Differ. 2002, 9, 1069–1077. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Uyttenhove, C.; Pilotte, L.; Théate, I.; Stroobant, V.; Colau, D.; Parmentier, N.; Boon, T.; Van den Eynde, B.J. Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2, 3-dioxygenase. Nat. Med. 2003, 9, 1269–1274. [Google Scholar] [CrossRef] [PubMed]
  108. Puccetti, P.; Grohmann, U. IDO and regulatory T cells: A role for reverse signalling and non-canonical NF-κB activation. Nat. Rev. Immunol. 2007, 7, 817–823. [Google Scholar] [CrossRef]
  109. Yang, C.; He, B.; Zheng, Q.; Wang, D.; Qin, M.; Zhang, H.; Dai, W.; Zhang, Q.; Meng, X.; Wang, X. Nano-encapsulated tryptanthrin derivative for combined anticancer therapy via inhibiting indoleamine 2, 3-dioxygenase and inducing immunogenic cell death. Nanomedicine 2019, 14, 2423–2440. [Google Scholar] [CrossRef] [PubMed]
  110. Apolonio, J.S.; de Souza Gonçalves, V.L.; Santos, M.L.C.; Luz, M.S.; Souza, J.V.S.; Pinheiro, S.L.R.; de Souza, W.R.; Loureiro, M.S.; de Melo, F.F. Oncolytic virus therapy in cancer: A current review. World J. Virol. 2021, 10, 229–255. [Google Scholar] [CrossRef]
  111. Kalus, P.; De Munck, J.; Vanbellingen, S.; Carreer, L.; Laeremans, T.; Broos, K.; Dufait, I.; Schwarze, J.K.; Van Riet, I.; Neyns, B.; et al. Oncolytic Herpes Simplex Virus Type 1 Induces Immunogenic Cell Death Resulting in Maturation of BDCA-1+ Myeloid Dendritic Cells. Int. J. Mol. Sci. 2022, 23, 4865. [Google Scholar] [CrossRef] [PubMed]
  112. Ingram, J.R.; Blomberg, O.S.; Sockolosky, J.T.; Ali, L.; Schmidt, F.I.; Pishesha, N.; Espinosa, C.; Dougan, S.K.; Garcia, K.C.; Ploegh, H.L.; et al. Localized CD47 blockade enhances immunotherapy for murine melanoma. Proc. Natl. Acad. Sci. USA 2017, 114, 10184–10189. [Google Scholar] [CrossRef] [PubMed]
  113. Jalil, A.R.; Andrechak, J.C.; Hayes, B.H.; Chenoweth, D.M.; Discher, D.E. Human CD47-Derived Cyclic Peptides Enhance Engulfment of mAb-Targeted Melanoma by Primary Macrophages. Bioconjug. Chem. 2022, 33, 1973–1982. [Google Scholar] [CrossRef] [PubMed]
  114. Zhang, B.; Shu, Y.; Hu, S.; Qi, Z.; Chen, Y.; Ma, J.; Wang, Y.; Cheng, P. In situ tumor vaccine expressing anti-CD47 antibody enhances antitumor immunity. Front. Oncol. 2022, 12, 897561. [Google Scholar] [CrossRef]
  115. Garofalo, M.; Pancer, K.W.; Wieczorek, M.; Staniszewska, M.; Salmaso, S.; Caliceti, P.; Kuryk, L. From Immunosuppression to Immunomodulation-Turning Cold Tumours into Hot. J. Cancer 2022, 13, 2884–2892. [Google Scholar] [CrossRef]
  116. Gleisner, M.A.; Pereda, C.; Tittarelli, A.; Navarrete, M.; Fuentes, C.; Ávalos, I.; Tempio, F.; Araya, J.P.; Becker, M.I.; González, F.E.; et al. A heat-shocked melanoma cell lysate vaccine enhances tumor infiltration by prototypic effector T cells inhibiting tumor growth. J. Immunother. Cancer 2020, 8, e000999. [Google Scholar] [CrossRef]
  117. Ott, P.A.; Hu-Lieskovan, S.; Chmielowski, B.; Govindan, R.; Naing, A.; Bhardwaj, N.; Margolin, K.; Awad, M.M.; Hellmann, M.D.; Lin, J.J.; et al. A phase Ib trial of personalized neoantigen therapy plus anti-PD-1 in patients with advanced melanoma, non-small cell lung cancer, or bladder cancer. Cell 2020, 183, 347–362.e24. [Google Scholar] [CrossRef]
  118. Malouff, T.D.; Mahajan, A.; Krishnan, S.; Beltran, C.; Seneviratne, D.S.; Trifiletti, D.M. Carbon ion therapy: A modern review of an emerging technology. Front. Oncol. 2020, 10, 82. [Google Scholar] [CrossRef] [Green Version]
  119. Particle Therapy Facilities in Clinical Operation. 2023. Available online: https://www.ptcog.site/index.php/facilities-in-operation-public (accessed on 7 March 2023).
  120. Zhou, H.; Tu, C.; Yang, P.; Li, J.; Kepp, O.; Li, H.; Zhang, L.; Zhang, L.; Zhao, Y.; Zhang, T.; et al. Carbon ion radiotherapy triggers immunogenic cell death and sensitizes melanoma to anti-PD-1 therapy in mice. Oncoimmunology 2022, 11, 2057892. [Google Scholar] [CrossRef]
  121. Zhou, H.; Yang, P.; Li, H.; Zhang, L.; Li, J.; Zhang, T.; Sheng, C.; Wang, J. Carbon ion radiotherapy boosts anti-tumour immune responses by inhibiting myeloid-derived suppressor cells in melanoma-bearing mice. Cell Death Discov. 2021, 7, 332. [Google Scholar] [CrossRef]
  122. Li, C.; Zhang, Q.; Li, Z.; Feng, S.; Luo, H.; Liu, R.; Wang, L.; Geng, Y.; Zhao, X.; Yang, Z.; et al. Efficacy and safety of carbon-ion radiotherapy for the malignant melanoma: A systematic review. Cancer Med. 2020, 9, 5293–5305. [Google Scholar] [CrossRef]
  123. Hou, Y.-J.; Yang, X.-X.; Liu, R.-Q.; Zhao, D.; Guo, C.-X.; Zhu, A.-C.; Wen, M.-N.; Liu, Z.; Qu, G.-F.; Meng, H.-X. Pathological mechanism of photodynamic therapy and photothermal therapy based on nanoparticles. Int. J. Nanomed. 2020, 15, 6827–6838. [Google Scholar] [CrossRef]
  124. Naidoo, C.; Kruger, C.A.; Abrahamse, H. Photodynamic therapy for metastatic melanoma treatment: A review. Technol. Cancer Res. Treat. 2018, 17, 1533033818791795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Heshmati Aghda, N.; Torres Hurtado, S.; Abdulsahib, S.M.; Lara, E.J.; Tunnell, J.W.; Betancourt, T. Dual photothermal/chemotherapy of melanoma cells with albumin nanoparticles carrying indocyanine green and doxorubicin leads to immunogenic cell death. Macromol. Biosci. 2022, 22, e2100353. [Google Scholar] [CrossRef] [PubMed]
  126. Song, R.; Li, T.; Ye, J.; Sun, F.; Hou, B.; Saeed, M.; Gao, J.; Wang, Y.; Zhu, Q.; Xu, Z.; et al. Acidity-activatable dynamic nanoparticles boosting ferroptotic cell death for immunotherapy of cancer. Adv. Mater. 2021, 33, e2101155. [Google Scholar] [CrossRef] [PubMed]
  127. Morais, J.A.V.; Almeida, L.R.; Rodrigues, M.C.; Azevedo, R.B.; Muehlmann, L.A. The induction of immunogenic cell death by photodynamic therapy in B16F10 cells in vitro is effected by the concentration of the photosensitizer. Photodiag. Photodyn. Ther. 2021, 35, 102392. [Google Scholar] [CrossRef]
  128. Konda, P.; Roque III, J.A.; Lifshits, L.M.; Alcos, A.; Azzam, E.; Shi, G.; Cameron, C.G.; McFarland, S.A.; Gujar, S. Photodynamic therapy of melanoma with new, structurally similar, NIR-absorbing ruthenium (II) complexes promotes tumor growth control via distinct hallmarks of immunogenic cell death. Am. J. Cancer Res. 2022, 12, 210–228. [Google Scholar]
  129. Podolska, M.J.; Shan, X.; Janko, C.; Boukherroub, R.; Gaipl, U.S.; Szunerits, S.; Frey, B.; Muñoz, L.E. Graphene-induced hyperthermia (GIHT) combined with radiotherapy fosters immunogenic cell death. Front. Oncol. 2021, 11, 664615. [Google Scholar] [CrossRef]
  130. Li, X.; Zhong, S.; Zhang, C.; Li, P.; Ran, H.; Wang, Z. MAGE-targeted gold nanoparticles for ultrasound imaging-guided phototherapy in melanoma. Biomed. Res. Int. 2020, 2020, 6863231. [Google Scholar] [CrossRef]
  131. Tatsuno, K.; Yamazaki, T.; Hanlon, D.; Han, P.; Robinson, E.; Sobolev, O.; Yurter, A.; Rivera-Molina, F.; Arshad, N.; Edelson, R.L.; et al. Extracorporeal photochemotherapy induces bona fide immunogenic cell death. Cell Death Dis. 2019, 10, 578. [Google Scholar] [CrossRef] [Green Version]
  132. Curley, C.T.; Sheybani, N.D.; Bullock, T.N.; Price, R.J. Focused ultrasound immunotherapy for central nervous system pathologies: Challenges and opportunities. Theranostics 2017, 7, 3608–3623. [Google Scholar] [CrossRef]
  133. Dauba, A.; Delalande, A.; Kamimura, H.A.; Conti, A.; Larrat, B.; Tsapis, N.; Novell, A. Recent advances on ultrasound contrast agents for blood-brain barrier opening with focused ultrasound. Pharmaceutics 2020, 12, 1125. [Google Scholar] [CrossRef]
  134. Kennedy, J.E. High-intensity focused ultrasound in the treatment of solid tumours. Nat. Rev. Cancer 2005, 5, 321–327. [Google Scholar] [CrossRef]
  135. Yuan, S.-M.; Li, H.; Yang, M.; Zha, H.; Sun, H.; Li, X.-R.; Li, A.-F.; Gu, Y.; Duan, L.; Luo, J.-Y.; et al. High intensity focused ultrasound enhances anti-tumor immunity by inhibiting the negative regulatory effect of miR-134 on CD86 in a murine melanoma model. Oncotarget 2015, 6, 37626–37637. [Google Scholar] [CrossRef]
  136. Wu, F.; Zhou, L.; Chen, W.R. Host antitumour immune responses to HIFU ablation. Int. J. Hyperth. 2007, 23, 165–171. [Google Scholar] [CrossRef] [PubMed]
  137. Xing, Y.; Lu, X.; Pua, E.C.; Zhong, P. The effect of high intensity focused ultrasound treatment on metastases in a murine melanoma model. Biochem. Biophys. Res. Commun. 2008, 375, 645–650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Marra, A.; Ferrone, C.R.; Fusciello, C.; Scognamiglio, G.; Ferrone, S.; Pepe, S.; Perri, F.; Sabbatino, F. Translational research in cutaneous melanoma: New therapeutic perspectives. Anti-Cancer Agents Med. Chem. 2018, 18, 166–181. [Google Scholar] [CrossRef]
  139. Singh, M.P.; Sethuraman, S.N.; Ritchey, J.; Fiering, S.; Guha, C.; Malayer, J.; Ranjan, A. In-situ vaccination using focused ultrasound heating and anti-CD-40 agonistic antibody enhances T-cell mediated local and abscopal effects in murine melanoma. Int. J. Hyperth. 2019, 36, 64–73. [Google Scholar] [CrossRef] [Green Version]
  140. Hu, J.; He, J.; Wang, Y.; Zhao, Y.; Fang, K.; Dong, Y.; Chen, Y.; Zhang, Y.; Zhang, C.; Wang, H.; et al. Ultrasound combined with nanobubbles promotes systemic anticancer immunity and augments anti-PD1 efficacy. J. Immunother. Cancer 2022, 10, e003408. [Google Scholar] [CrossRef]
  141. Prasad, C.; Banerjee, R. Ultrasound-triggered spatiotemporal delivery of topotecan and curcumin as combination therapy for cancer. J. Pharm. Exp. Ther. 2019, 370, 876–893. [Google Scholar] [CrossRef] [PubMed]
  142. Feril, L.B., Jr.; Yamaguchi, K.; Ikeda-Dantsuji, Y.; Furusawa, Y.; Tabuchi, Y.; Takasaki, I.; Ogawa, R.; Cui, Z.-G.; Tachibana, K. Low-intensity ultrasound inhibits melanoma cell proliferation in vitro and tumor growth in vivo. J. Med. Ultrason. 2021, 48, 451–461. [Google Scholar] [CrossRef] [PubMed]
  143. Campana, L.G.; Peric, B.; Mascherini, M.; Spina, R.; Kunte, C.; Kis, E.; Rozsa, P.; Quaglino, P.; Jones, R.P.; Clover, A.J.P.; et al. Combination of pembrolizumab with electrochemotherapy in cutaneous metastases from melanoma: A comparative retrospective study from the InspECT and Slovenian Cancer Registry. Cancers 2021, 13, 4289. [Google Scholar] [CrossRef] [PubMed]
  144. Gorgizadeh, M.; Azarpira, N.; Lotfi, M.; Daneshvar, F.; Salehi, F.; Sattarahmady, N. Sonodynamic cancer therapy by a nickel ferrite/carbon nanocomposite on melanoma tumor: In vitro and in vivo studies. Photodiag. Photodyn. Ther. 2019, 27, 27–33. [Google Scholar] [CrossRef]
  145. Sheehan, D.; Sheehan, K.; Sheehan, J. Sonodynamic therapy for metastatic melanoma to the brain. J. Neuro-Oncol. 2021, 153, 373–374. [Google Scholar] [CrossRef] [PubMed]
  146. Do, A.-V.; Geary, S.M.; Seol, D.; Tobias, P.; Carlsen, D.; Leelakanok, N.; Martin, J.A.; Salem, A.K. Combining ultrasound and intratumoral administration of doxorubicin-loaded microspheres to enhance tumor cell killing. Int. J. Pharm. 2018, 539, 139–146. [Google Scholar] [CrossRef]
  147. Tamura, Y.; Ito, A.; Wakamatsu, K.; Kamiya, T.; Torigoe, T.; Honda, H.; Yamashita, T.; Uhara, H.; Ito, S.; Jimbow, K. Immunomodulation of melanoma by chemo-thermo-immunotherapy using conjugates of melanogenesis substrate NPrCAP and magnetite nanoparticles: A review. Int. J. Mol. Sci. 2022, 23, 6457. [Google Scholar] [CrossRef]
  148. Nishikawa, A.; Suzuki, Y.; Kaneko, M.; Ito, A. Combination of magnetic hyperthermia and immunomodulators to drive complete tumor regression of poorly immunogenic melanoma. Cancer Immunol. Immunother. 2022, 72, 1493–1504. [Google Scholar] [CrossRef]
  149. Pakhomova, O.N.; Gregory, B.W.; Semenov, I.; Pakhomov, A.G. Two modes of cell death caused by exposure to nanosecond pulsed electric field. PLoS ONE 2013, 8, e70278. [Google Scholar] [CrossRef] [Green Version]
  150. Beebe, S.J.; Fox, P.M.; Rec, L.J.; Willis, L.K.; Schoenbach, K.H. Nanosecond, high-intensity pulsed electric fields induce apoptosis in human cells. FASEB J. 2003, 17, 1493–1495. [Google Scholar] [CrossRef] [Green Version]
  151. Lin, A.; Gorbanev, Y.; De Backer, J.; Van Loenhout, J.; Van Boxem, W.; Lemière, F.; Cos, P.; Dewilde, S.; Smits, E.; Bogaerts, A. Non-thermal plasma as a unique delivery system of short-lived reactive oxygen and nitrogen species for immunogenic cell death in melanoma cells. Adv. Sci. 2019, 6, 1802062. [Google Scholar] [CrossRef] [Green Version]
  152. Azzariti, A.; Iacobazzi, R.M.; Di Fonte, R.; Porcelli, L.; Gristina, R.; Favia, P.; Fracassi, F.; Trizio, I.; Silvestris, N.; Guida, G. Plasma-activated medium triggers cell death and the presentation of immune activating danger signals in melanoma and pancreatic cancer cells. Sci. Rep. 2019, 9, 4099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Rossi, A.; Pakhomova, O.N.; Pakhomov, A.G.; Weygandt, S.; Bulysheva, A.A.; Murray, L.E.; Mollica, P.A.; Muratori, C. Mechanisms and immunogenicity of nsPEF-induced cell death in B16F10 melanoma tumors. Sci. Rep. 2019, 9, 431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Sagwal, S.K.; Pasqual-Melo, G.; Bodnar, Y.; Gandhirajan, R.K.; Bekeschus, S. Combination of chemotherapy and physical plasma elicits melanoma cell death via upregulation of SLC22A16. Cell Death Dis. 2018, 9, 1179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Zheng, Y.; Delgoffe, G.M.; Meyer, C.F.; Chan, W.; Powell, J.D. Anergic T cells are metabolically anergic. J. Immunol. 2009, 183, 6095–6101. [Google Scholar] [CrossRef] [Green Version]
  156. Seiwert, T.Y.; Kiess, A.P. Time to Debunk an Urban Myth? The “Abscopal Effect” with Radiation and Anti-PD-1. J. Clin. Oncol. 2021, 39, 1–3. [Google Scholar] [CrossRef]
  157. Bandyopadhyay, S.; Quinn, T.J.; Scandiuzzi, L.; Basu, I.; Partanen, A.; Tomé, W.A.; Macian, F.; Guha, C. Low-intensity focused ultrasound induces reversal of tumor-induced T cell tolerance and prevents immune escape. J. Immunol. 2016, 196, 1964–1976. [Google Scholar] [CrossRef] [Green Version]
  158. Skalina, K.A.; Singh, S.; Chavez, C.G.; Macian, F.; Guha, C. Low intensity focused ultrasound (LOFU)-mediated acoustic immune priming and ablative radiation therapy for in situ tumor vaccines. Sci. Rep. 2019, 9, 15516. [Google Scholar] [CrossRef] [Green Version]
  159. Heissmeyer, V.; Macián, F.; Im, S.-H.; Varma, R.; Feske, S.; Venuprasad, K.; Gu, H.; Liu, Y.-C.; Dustin, M.L.; Rao, A. Calcineurin imposes T cell unresponsiveness through targeted proteolysis of signaling proteins. Nat. Immunol. 2004, 5, 255–265. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of ICD and its effect on the anti-tumor immune response. After exposure to a stressor or ICD-inducer, tumor cells experience ER stress and subsequently upregulate the UPR. One of the three arms of the UPR which involves PERK has been mechanistically implicated in the induction of ICD and is also associated with the regulation of autophagy through ATF4. Upregulation of the UPR may lead to various forms of ICD, characterized by the translocation of calreticulin (CRT) from the ER to the plasma membrane and the release of DAMPs including HMGB1 and ATP. Immature DCs phagocytose soluble tumor antigens and/or dying tumor cells with ecto-CRT “eat me” signals. Tumor cells undergoing ICD also release type-1 interferons (IFN1), essential drivers of anti-tumor immunity. The released DAMPs interact with TLRs on DCs within the tumor microenvironment, leading to the maturation and activation of DCs with increased expression of MHC-II and the co-stimulatory molecules CD80 and CD86. Mature DCs migrate to the draining lymph node and present tumor antigens via both class II and class I MHC pathways to prime and activate CD4+ and CD8+ T-cells, respectively. CD8+ T-cells are the primary effectors of the anti-tumor adaptive immune response and are able to directly kill tumor cells using perforin and granzyme B.
Figure 1. Schematic representation of ICD and its effect on the anti-tumor immune response. After exposure to a stressor or ICD-inducer, tumor cells experience ER stress and subsequently upregulate the UPR. One of the three arms of the UPR which involves PERK has been mechanistically implicated in the induction of ICD and is also associated with the regulation of autophagy through ATF4. Upregulation of the UPR may lead to various forms of ICD, characterized by the translocation of calreticulin (CRT) from the ER to the plasma membrane and the release of DAMPs including HMGB1 and ATP. Immature DCs phagocytose soluble tumor antigens and/or dying tumor cells with ecto-CRT “eat me” signals. Tumor cells undergoing ICD also release type-1 interferons (IFN1), essential drivers of anti-tumor immunity. The released DAMPs interact with TLRs on DCs within the tumor microenvironment, leading to the maturation and activation of DCs with increased expression of MHC-II and the co-stimulatory molecules CD80 and CD86. Mature DCs migrate to the draining lymph node and present tumor antigens via both class II and class I MHC pathways to prime and activate CD4+ and CD8+ T-cells, respectively. CD8+ T-cells are the primary effectors of the anti-tumor adaptive immune response and are able to directly kill tumor cells using perforin and granzyme B.
Biomedicines 11 02245 g001
Table 1. Current FDA-approved systemic melanoma therapies.
Table 1. Current FDA-approved systemic melanoma therapies.
TherapyGeneric Name (Brand Name)References
Checkpoint inhibitor therapiesNivolumab and Relatlimab Combination Therapy (Opdualag)[3]
Ipilimumab (Yervoy)[4,5]
Pembrolizumab (Keytruda)[6]
Nivolumab (Opdivo)[7]
BRAF/MEK inhibitorsBinimetinib (Mektovi) + Encorafenib (Braftovi)[8]
Vemurafenib (Zelboraf) + Cobimetinib (Cotellic)[9]
Dabrafenib (Tafinlar) + Trametinib (Mekinist)[10,11]
Cytotoxic chemotherapyDacarbazine[12]
Other immunotherapiesInterleukin-2 (Aldesleukin, Proleukin)[13]
Recombinant Interferon Alfa-2b (Intron A)[14]
Table 2. Targeted ICD-inducers and their mechanisms of action.
Table 2. Targeted ICD-inducers and their mechanisms of action.
ICD-InducerExamplesMechanismReferences
Chemical/small molecule inducersChemotherapeutics
(i.e., doxorubicin and paclitaxel)
Increase tumor cell expression of PD-L1[87]
Celastrol
(quinone methide family)
Induces tumor cell autophagy; decreases tumor cell expression of PD-L1 [88]
Chromomycins A5-8
(antibiotics)
Induces tumor cell apoptosis[89]
PolyphenolsInduces PERK arm of the UPR[90]
RT53
(peptide)
Promotes B16-F10 tumor regression [91]
Oncolytic virusesTalimogene laherparepvec (T-VEC)Oncolytic herpes simplex virus HSV-1 that infects tumor cells and encodes granulocyte-macrophage colony stimulating factor;
Enhances CD8+ T-cell recruitment via STING-mediated pathway;
Modulation to express GALV-GP-R- leads to enhanced ICD and abscopal effect
[92,93,94]
FixVac (BNT111)Liposomal RNA vaccine (Clinical Trial NCT02410733) that enhances anti-tumor immune response when used with other ICD-inducers [95]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pelka, S.; Guha, C. Enhancing Immunogenicity in Metastatic Melanoma: Adjuvant Therapies to Promote the Anti-Tumor Immune Response. Biomedicines 2023, 11, 2245. https://doi.org/10.3390/biomedicines11082245

AMA Style

Pelka S, Guha C. Enhancing Immunogenicity in Metastatic Melanoma: Adjuvant Therapies to Promote the Anti-Tumor Immune Response. Biomedicines. 2023; 11(8):2245. https://doi.org/10.3390/biomedicines11082245

Chicago/Turabian Style

Pelka, Sandra, and Chandan Guha. 2023. "Enhancing Immunogenicity in Metastatic Melanoma: Adjuvant Therapies to Promote the Anti-Tumor Immune Response" Biomedicines 11, no. 8: 2245. https://doi.org/10.3390/biomedicines11082245

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop