Next Article in Journal
Regulation of Conidiogenesis in Aspergillus flavus
Next Article in Special Issue
Cell-Free DNA Sequencing Reveals Gene Variants in DNA Damage Repair Genes Associated with Prognosis of Prostate Cancer Patients
Previous Article in Journal
Botanicals and Oral Stem Cell Mediated Regeneration: A Paradigm Shift from Artificial to Biological Replacement
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Boosting the Immune Response—Combining Local and Immune Therapy for Prostate Cancer Treatment

1
University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
2
Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wrocław, Poland
3
Department of Drugs Form Technology, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wrocław, Poland
*
Authors to whom correspondence should be addressed.
Cells 2022, 11(18), 2793; https://doi.org/10.3390/cells11182793
Submission received: 24 July 2022 / Revised: 3 September 2022 / Accepted: 6 September 2022 / Published: 7 September 2022
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Prostate Cancer)

Abstract

:
Due to its slow progression and susceptibility to radical forms of treatment, low-grade PC is associated with high overall survival (OS). With the clinical progression of PC, the therapy is becoming more complex. The immunosuppressive tumor microenvironment (TME) makes PC a difficult target for most immunotherapeutics. Its general immune resistance is established by e.g., immune evasion through Treg cells, synthesis of immunosuppressive mediators, and the defective expression of surface neoantigens. The success of sipuleucel-T in clinical trials initiated several other clinical studies that specifically target the immune escape of tumors and eliminate the immunosuppressive properties of the TME. In the settings of PC treatment, this can be commonly achieved with radiation therapy (RT). In addition, focal therapies usually applied for localized PC, such as high-intensity focused ultrasound (HIFU) therapy, cryotherapy, photodynamic therapy (PDT), and irreversible electroporation (IRE) were shown to boost the anti-cancer response. Nevertheless, the present guidelines restrict their application to the context of a clinical trial or a prospective cohort study. This review explains how RT and focal therapies enhance the immune response. We also provide data supporting the combination of RT and focal treatments with immune therapies.

1. Introduction

In 2020, prostate cancer (PC) was the second most frequent cancer and the fifth cause of cancer-related death among men. In more than half of the countries in the world, it was the most frequently diagnosed cancer in men [1]. While mortality rates are relatively low in comparison to other malignancies, metastatic castration-resistant prostate cancer (mCRPC) remains an incurable condition, with few treatment strategies providing any clinical benefit [2].
Focal therapies are minimally invasive treatment strategies used in the management of PC to provide local control of the disease, minimalizing the risk of possible complications. The first prospective clinical trials showed promising local disease control, especially with a short-term perspective [3]. The immunological impact of focal therapies, as well as the immunotherapy of PC itself, have been addressed by academic research for years. Thus, it was a substantial breakthrough when sipuleucel-T became the first therapeutic vaccine for patients with mCRPC approved by the United States Food and Drug Administration (FDA) and the first autologous cellular therapeutic vaccine in oncology [4,5]. Nevertheless, the clinical benefit of immunotherapy alone remains limited partially due to low-grade inflammation in the tumor microenvironment (TME), as well as uncontrollable cell divisions and the suppressed expression of tumor antigens [6,7]. However, both radiotherapy (RT) and various focal therapies have the potential to activate the anti-tumor immune response and, therefore, enhance the efficacy of immunotherapy [8,9,10].
The major purposes of this non-systematic review were to identify the immune effect of RT and focal therapies, including high-intensity focused ultrasound (HIFU), cryotherapy, photodynamic therapy (PDT), and irreversible electroporation (IRE), and to compile available knowledge on different combinational therapies including both a focal and an immunotherapeutic component. To provide background data on the immunology of PC, we thoroughly described the TME and various immune evasion mechanisms. Furthermore, for better understanding of the current status of immunotherapy, we summarized the most recent scientific data on different management options in PC, including immune checkpoint inhibitors (ICIs) and cancer vaccines.

2. Data Acquisition

For the purposes of this non-systematic review, we conducted a comprehensive English language literature search for original articles, meta-analyses and reviews using PubMed and grey literature through June and July 2022. We searched for various combinations of the following terms: prostate cancer; immunotherapy; focal therapy; cancer vaccine; combination therapy; tumor microenvironment. We found 1669 related articles, and the final number of papers selected for this manuscript was 392. Studies with the highest level of evidence and relevance to the discussed topics (275) were selected, with the consensus of the authors.

3. Immunological Background of Prostate Cancer

3.1. Prostate Cancer Microenvironment

The microenvironment of PC consists of numerous elements, including both neoplastic cells and diverse host cells. The host component comprises stromal cells, the extracellular matrix, endothelial and vascular cells, immune cells, and various soluble factors, such as IL-6 and receptor activator of nuclear factor kappa-B ligand (RANKL) [11]. The TME in PC plays an ambiguous role in carcinogenesis. Particularly, the impact of the immune system is highly complex, as both innate and adaptive immune response mechanisms can provide anti-neoplastic activity, as well as propagate carcinogenesis [12]. For example, cytotoxic T lymphocytes (CTLs), one of the most important cancer cell killers, secrete transforming growth factor-beta (TGF-β), which both supports tumor growth and induces immune suppression [13].
There is a multitude of mechanisms affecting the TME in PC and contributing to immunosuppression, including the inhibition of neoantigen expression, instability of rapid cell division, DNA damage response (DDR) gene defects, decreased human leucocyte antigen (HLA) expression, phosphatase and tensin homolog (PTEN) protein loss, and dysfunction of interferon (IFN) type I signaling [7].

3.2. T Cell Infiltration

Many immune cell types play a role in TME functionality, although the T cell population, especially CTLs, are considered most vital [14]. They are the key elements of the physiological cancer immunity cycle, which is briefly summarized in Figure 1 [15]. T cells are recruited from peripheral blood after antigen-presenting cells (APCs), specifically dendritic cells (DCs) and macrophages, capture neoantigens released by the tumor. Presenting the abovementioned antigens to CTLs using a major histocompatibility complex (MHC) is called priming and takes place in the local lymph nodes. This results in recruiting and stimulating more T cells, including CD4+ cells. CTLs infiltrate the tumor, recognize cancer cells, and kill them. Neoantigens are then released and the process comes full circle [15]. The localization and density of tumor infiltrating lymphocytes (TILs) and memory T cells within the center of the tumor and its margins were the foundation for creating the “immunoscore”. It divides tumors into two groups: T cell inflamed (“hot”) and non-T cell inflamed (“cold”) [16]. This immune contexture is significant in the efficacy of therapy in a variety of cancers. Many publications indicate that a high level of TILs shows a positive prognostic value [17,18,19,20,21,22]. PC is primarily described as a “cold” tumor, with a low inflammation burden and immune activation [23]. However, the impact of the TME on PC oncological outcomes is unclear [7]. Some studies show that the high intratumoral density of CTLs is associated with improved cancer-specific survival (CSS) in PC patients undergoing RP [24,25]. Others show that the higher the level of CTL infiltration in PC, the greater the risk of distant metastases and biochemical recurrence [12,26]. Although the connection between inflammation and tumorigenesis remains unclear, one of the main goals of various local pre-immunotherapy techniques is to propagate the inflammation of the TME, converting it to inflamed and susceptible to immunotherapy [27].

3.3. Regulation of the T Cell Response

After a T cell is initially activated during priming, the second step of activation takes place: binding of the costimulatory molecules, CD80 (B7-1) or CD86 (B7-2), which serve as ligands on APCs, and CD28, a receptor expressed on T cells [28,29,30]. The cytotoxic T lymphocyte antigen 4 (CTLA-4 or CD152) is a co-inhibitory glycoprotein receptor expressed on the surface of the T cell, competing with CD28 for B7 ligands. CTLA-4 is induced after T cell activation (except for regulatory T cells (Tregs), which continuously express it), and due to its higher affinity for B7 molecules, it successfully outcompetes the CD28 receptor [31,32,33,34]. The B7:CTLA4 interaction leads to inhibition of the cell cycle progression through IL-2 accumulation [35,36]. Programmed death receptor 1 (PD-1) is another co-inhibitory receptor on the surface of T- and B cells. PD-1 ligand 1 (PD-L1 or CD274) and PD-1 ligand 2 (PD-L2 or CD273) are two known ligands for the PD-1 receptor, expressed on macrophages, DCs, and other immune cells [37]. Although the interaction of PD-L2 and PD-1 also has an immunosuppressive outcome, it is the PD-L1:PD-1 binding that induces the conversion of naïve T cells into Tregs [38,39,40,41]. CTLA-4, PD-1, and its ligands are parts of the B7 superfamily of molecules and are the most vital immune checkpoints (ICPs) [42].

3.4. Immune Evasion Mechanisms

Cancer cells have developed several immune evasion mechanisms associated with TME components. Immune evasion may be described as the entirety of biochemical interactions leading to the suppression of the natural immune response to tumor cells. The spectrum of possible “back doors” can be generally divided into a few mechanisms. These include: (1) immune evasion through immune cells (most notably Tregs), (2) synthesis of immune-suppressive mediators, and (3) defective expression of surface neoantigens [43]. Additionally, important contributors to the TME, affecting cancer progression and the response to therapy, are hypoxia and glucose restriction, which have already been described in a multitude of malignancies [44,45,46].

3.4.1. The Role of Specific Immune Cells

One of the cancer immune evasion mechanisms is CD4+ CD25+ FOXP3+ Tregs activity, as its physiological role is to modulate effector T cells to support immunological tolerance to self-antigens (self-Ags) [47,48,49]. Tregs drawn by the tumor have higher suppressive properties compared to circulating Tregs and are able to inhibit the anti-tumor activity of other immune cells directly using cell–cell interactions or indirectly through synthesis and the secretion of mediators, e.g., TGF-β, interleukin 10 (IL-10) [50,51]. Many tumor-associated Ags are expressed by host cells and can therefore act like self-Ags, which further emphasizes the role of Tregs in immune evasion [52,53].
Myeloid-derived suppressor cells (MDSCs) are another heterogeneous group comprising immature DCs, granulocytes, and macrophages. Overproduction and concentration of these cell types in an inflammatory environment are correlated with the immunosuppressive qualities of the TME [54,55]. Their functions include the inhibition of CLTs through various mechanisms (e.g., producing reactive oxygen species (ROS) or interactions with the T cell receptor [TCR]), suppressing natural killer (NK) cells, and Tregs induction [56,57,58,59,60]. The level of MDSCs correlates with the stage of PC and applied treatment, as well as with the serum levels of crucial inflammatory mediators—IL-6 and IL-8 [61,62,63].
DCs are the most efficient APCs, but their functionality is mutilated due to the modulatory activity of tumors. Impaired DCs have lower levels of CD80, CD86, and CD40, thus they cannot present antigens and activate T cells effectively enough [43,64,65]. The role of CD40 is highly complex as it connects the T- and B cell responses. Namely, when DCs remain active and secrete IL-12, they may interact with CD40L on both T cells and B cells [66,67,68,69]. The first interaction induces the Th1 and IFN-γ secretion by the T cells, and the second induces the class switching between IgG and IgA in B cells [70,71,72]. In addition, the reciprocal expression of CD40 and CD40L on DCs, T cells, and B cells links the humoral and cellular immune response, thus the reduced level of CD40 might lead to impairments in both responses [72].
Tumor-associated macrophages (TAMs) are another important group contributing to the PC TME. TAMs, especially the M2 type, can stimulate tumor growth through the secretion of various mediators such as TGF-β, IL-10, and vascular endothelial growth factor (VEGF) [43,73,74,75]. Overexpression of TAMs in PC is correlated with unfavorable oncological outcomes in patients with PC, including biochemical recurrence (BCR), or worse, distant metastasis-free survival [76,77,78].
Furthermore, increased concentration of TAMs is observable in metastatic lymph nodes in comparison to the primary prostate tumor [79,80]. Shortage of available data on other TME differences in primary and metastatic PC prevents us from presenting a thorough comparison, but it is worth mentioning that this immunological heterogeneity has already been described in other malignancies, including breast, lung, colorectal cancers, and brain tumors [81,82,83,84,85,86,87].
The role of B cells in immune evasion is not well understood in the case of PC. However, B cell infiltration has prognostic significance in different cancers such as breast cancer and melanoma [88]. B cell TILs secrete a significant member of the TNF family, lymphotoxin (LT), which promotes survival and proliferation of androgen-deprived cells, therefore encouraging castration-resistant PC (CRPC) development [89].

3.4.2. Immunosuppressive Mediators

There are many immunosuppressive cytokines which aid tumors in immune evasion through the promotion of tumor proliferation, chemoresistance, angiogenesis, and migration, and these are most notably TGF-β, VEGF, IL-6, RANKL, and the CXCL family [90]. TGF-β is one of the most vital mediators, acting both as a direct growth-promoting factor, as well as a stimulator of CD4+ T cells–Tregs transformation [91,92,93]. Its other roles include promoting angiogenesis, downregulating HLA-1 expression, and inducing epithelial–mesenchymal transition (EMT) [90,94,95,96,97]. Another important cytokine is VEGF, which also contributes to tumor growth, as well as inhibiting DC differentiation. A similar role is performed by cancer-associated ganglioside antigens, which conduct an immunosuppressive activity through impairing CTLs and DCs [43,98,99].

3.4.3. Dysfunctional Expression of Surface Neoantigens

MHC Class I proteins are found on nucleated cells and platelet surfaces, and their role is to be recognized by CD8+ T cells, which trigger the immune response against certain antigens by activating T cells and leading to target cell destruction [5,100,101]. Decreased MHC I presentation of tumor-associated antigens is one of the immune evasion mechanisms of PC [102,103]. A study by Yitalo et al. revealed heterogeneity in MHC I expression in PC. Bone metastases showed noticeably lower MHC I expression in comparison to the primary tumor. Moreover, a subgroup of metastases showed high MHC I expression, while the majority of metastases showed a decrease. These findings indicate that there are molecular subtypes of PC which may be less responsive to immunotherapy [104].

3.4.4. Monitoring of the Immune Response

The immune effects provided by immunotherapy or initiated by focal therapies are difficult to unequivocally assess. There is no gold standard of immunological activity, and various studies focus on different aspects of induced immunomodulation. Some of the most valid indicators of this state include: immunogenic cell death; concentration of CTLs, DCs, and other immune cells; the expression level of certain antigens and factors (e.g., PD-L1, MHC, IL-1, IL-6, interferons); and the abscopal effect [105,106,107]. Nevertheless, a well-designed randomized clinical trial comparing the immune effects of the abovementioned treatment strategies would be vital to adequately evaluate the immunomodulatory effects provided by focal therapies.

4. Immunotherapeutic Options for Prostate Cancer

4.1. Immune Checkpoint Inhibitors

ICIs are novel treatment options gaining more and more interest as they already appear to act as successful strategies in cancers such as melanoma and lung cancer [108,109,110,111,112,113,114]. Among CTLA-4 inhibitors used in oncology, there are ipilimumab and tremelimumab, while the most pivotal PD-1 inhibitors comprise nivolumab and pembrolizumab; atezolizumab, avelumab, and durvalumab belong to PD-L1 inhibitors [115,116,117].
There have been a few major studies on ICI efficacy in PC management in recent years; most trials investigated various immunotherapy combinations, especially with other immunotherapeutics or with systemic chemotherapy, or ADT. The preliminary results of the study by Sharma et al. (CheckMate 650, NCT02985957) showed promising anti-tumor activity of the nivolumab and ipilimumab combination in patients with mCRPC. The authors reported 25% objective response rate (ORR) in the pre-chemotherapy arm, and 10% ORR in the post-chemotherapy arm, as well as 5.5 and 3.8 months median radiographic progression-free survival (PFS), and 19.0 and 15.2 months median OS. However, research expansion is necessary to further assess the clinical outcomes of this therapy [118]. In the randomized phase III trial by Powles et al. (The IMbassador250 trial, NCT03016312), the combination of atezolizumab and enzalutamide failed to meet the endpoint of improved OS. However, the authors pointed out that proper patient selection may be vital to identify subgroups of patients who are susceptible to this therapy [119]. The phase III randomized study by Agarwal et al. (CONTACT-02, NCT04446117) will evaluate the combination of atezolizumab and cabozantinib vs. second novel hormone therapy (abiraterone or enzalumatide) in patients with mCRPC [120].
The European Association of Urology (EAU) guidelines indicate that pembrolizumab may be a valuable additional management strategy for mCRPC patients with high microsatellite instability [121,122]. This further suggests that the increase in immunotherapy efficacy in PC may require meticulous patient selection.
Immune checkpoint blockade in PC remains a poor monotherapeutic tool [123,124]. Among the reasons for this is the low level of T cell infiltration, the “cold” immunogenic profile of the tumor, mutational burden, and immune evasion mechanisms [125,126,127]. Thus, the interest in boosting the immune response, using other therapies beforehand, is now rapidly growing. It is believed that various therapy combinations can subdue tumor resistance to ICI therapy [128]. Studies evaluating ICI combinations with each other and with focal therapies show more promising results in comparison to monotherapeutic approaches [118,123]. However, the study by Palmer et al. revealed that the combination of ICIs with various other strategies (e.g., other immunotherapy, chemotherapy) shows no enhancement (additivity or synergy) in comparison to ICIs alone in 13 phase III clinical trials [129].
For the purpose of this review, we present results of various studies on the combination of ICIs and focal therapies in the sections concerning specific local treatment strategies.

4.2. Cancer Vaccines

Cancer vaccines usually consist of specific neoantigens and adjuvants that boost the immune response against these antigens. Their role is to support the adaptive immune system of the patient in neutralizing cancer cells [130]. Cancer vaccinations have already been combined with RT and focal therapies, due to their immunomodulatory effect, in various clinical trials, evaluating their efficacy in numerous malignancies including PC, melanoma, pancreatic, and lung cancers [131,132,133,134,135,136,137,138,139,140,141].

4.2.1. Dendritic Cell Vaccines—Sipuleucel-T, DCvac/PCa, and Others

As mentioned above, DCs are one of the most important features of the immune system; they are the most efficient APCs, which are not only able to activate T cells (both Tregs and CTLs) but also NK cells. DC vaccines require blood-derived DCs, pulsing them ex vivo with the tumor-associated antigen, activating them using the specific adjuvant, and then reinjecting them into the patient [142]. The first DC vaccine approved by the U.S. Food and Drug Administration (FDA) was sipuleucel-T (Provenge®), and so far, it remains the only DC vaccine for mCRPC [143,144]. Sipuleucel-T promotes an immune response against tumor cells using prostatic acid phosphatase (PAP) antigen-activated DCs [42]. A double-blind, placebo-controlled, multicenter phase III trial compared this DC vaccine to the placebo group, with the results of a 22% reduction in the risk of death and more than 4 months of improvement in overall survival (OS) [4]. Another trial showed even greater improvement in OS (up to 8.1 months), if sipuleucel-T therapy is extended by APC8015F, a variant of the DC vaccine prepared from cryopreserved cells which were frozen for future use [145].
DC vaccines are a very promising therapeutic tool, although they require further clinical trials and more attempts to combine them using different approaches [146]. There is only one ongoing trial assessing the combination of sipuleucel-T and other therapies: sipuleucel-T plus stereotactic ablative body radiation (SABR) (NCT01818986, phase II). A different phase III trial evaluates the efficiency of sipuleucel-T in reducing the progression of CRPC. The study includes active surveillance patients (the ProVent Study; NCT03686683).
DCvac/PCa is an autologous DC-based vaccine, in which DCs are pulsed with killed lymph node carcinoma of prostate (LNCaP) cells. Several clinical trials have investigated its efficacy in PC. However, only a few of them examine its combinations with focal therapies or RT. Fucikova et al. assessed the DCvac/PCa impact on PSA in patients with rising PSA after RP or salvage RT. PSA doubling time was significantly elongated in this variant [147]. Although the DCvac/PCa immunological impact is quite well documented, its translation to clinical benefits is needed, and further clinical trials are required, especially concerning different combinations of therapies. A recent clinical phase III trial (the Viable) by Vogelzang et al. investigated the DCvac/PCa combination with docetaxel and prednisone. The therapy failed to improve OS in patients with mCRPC [148].
Other DC-based vaccines that have been tested in the last decade in PC patients are prostate-specific membrane antigen (PSMA) and the survivin-loaded DC vaccine, mucin 1 (MUC1) vaccine, and the T cell receptor ɣ alternate reading frame protein (TARP) vaccine [149,150,151,152].

4.2.2. PROSTVAC—A PSA-Based Viral Vector Vaccine

One of the trailblazing PC vaccines is PROSTVAC (PSA-TRICOM), which comprises two recombinant poxvirus vectors containing transgenes for PSA and three costimulatory molecules: B7.1, ICAM-1, and LFA-3 [153,154]. A phase II trial analyzing neoadjuvant PROSTVAC in patients awaiting RP showed an increase in CD4+ and CD8+ T cell infiltration of the tumor, as well as a peripheral immune response to neoantigens in 13 out of the 25 patients [155]. However, this promising immune response does not yet translate into a clinical advantage. In a phase III trial, Gulley et al. concluded that despite the fact that the therapy was well-tolerated and safe for patients, treatment disappointingly showed no impact on median OS and alive without event (AWE) in patients with mCRPC [156]. Parsons et al. evaluated the preventive value of PROSTVAC in patients with localized PC which is managed by an active surveillance strategy. Although some initial data on the immunological effect of the vaccine are already available, we are looking forward to the summary of this phase II trial in the future (NCT02326805) [157].
Several ongoing clinical trials are investigating different combinational management strategies including PROSTVAC. These are evaluating, among others, the combination with nivolumab (NCT02933255, phase I/II) or nivolumab and ipilimumab (NCT03532217, phase I), with CV301 (a poxviral vaccine) and M7824 (a protein targeting PD-L1 and TGF-β) (NCT03315871, phase II), docetaxel (NCT02649855, phase II), or enzalutamide (NCT01867333, phase II).
TroVax is another viral vector, 5T4 (oncofetal glycoprotein) targeting vaccination. It is characterized by a good immune response in mCRPC and the potential to efficiently combine with docetaxel [158,159].

4.2.3. Peptide-Based Vaccines

Among peptide-based vaccines, one of the most interesting is GX301, consisting of four telomerase peptides and two adjuvants—Montanide ISA-51 and Imiquimod. Fenoglio et al. assessed its potential in a phase I/II clinical trial, revealing its immunological response in PC and renal cell cancer (RCC). An increase in PFS and OS were also observed [160]. Filaci et al. evaluated GX301 efficiency and immunological impact in mCRPC. The therapy did not increase OS, although they observed that a higher number of drug administrations was correlated with an increased immunological response [161].
Cell division-associated 1 (CDCA1) peptide vaccination was a topic of research in a phase I clinical trial by Obara et al. CDCA1 is a peptide overexpressed in a few malignancies, including PC. The authors indicated that the vaccine is well-tolerated, and it boosts the immunological response in patients with CRPC [162,163].
Other peptide-based vaccinations include the personalized peptide vaccination (PPV), which includes the administration of different HLA-matched peptides, multi-peptide vaccines, and a vaccine targeting the Ras homolog gene family member C (RhoC vaccine, RV 001, or onilcamotide) [164,165,166]. However, a recent phase IIb study of RhoVac (RV 001) failed to show its superiority over the placebo in PC (BRaVac, NCT04114825). Further phase II and III trials investigating various peptide-based vaccinations are required in the future.

4.2.4. Whole Tumor Cell Vaccines

GVAX is a vaccine consisting of genetically modified PC cells, which have undergone radiation. Studies suggest that this vaccination induces immune response through the activation of DCs and MDSCs [167]. A combinational therapy with ipilimumab has been investigated in a phase I trial by van den Eertwegh et al., which showed that GVAX is well-tolerated and safe for patients with mCRPC [168]. Once again, further clinical trials are required [169].

5. Focal Ablation and Immune Therapy Combination

5.1. High-Intensity Focused Ultrasound

Recently, HIFU appeared as a potential neoadjuvant-like therapy, serving as the first step of immunotherapeutic treatment. HIFU itself has already made an appearance in guidelines concerning PC treatment options, although only as an investigational therapeutic tool or as salvage therapy [170]. Essentially, this focal management strategy uses highly focused ultrasound waves to ablate cancer cells and has been investigated extensively in PC patients in a multitude of clinical trials since the late 1990s [171,172,173,174,175,176,177,178,179,180,181]. The most important benefit of HIFU is that it is minimally invasive when compared to surgical treatment, and it is devoid of systemic toxicity in comparison with androgen deprivation therapy (ADT) or chemotherapy. Nevertheless, possible adverse effects may quite frequently occur, and they include erectile dysfunction, urinary tract infections, rectal injuries, and more [182,183]. The properties of HIFU can be divided into a few groups—ablative and non-ablative (mechanical), immune, and biological effects. Induced activity depends on a multitude of factors including frequency, pressure, duty cycle, treatment time, achieved temperature, tissue susceptibility, and more. This allows us to distinguish several possible technique variants, such as thermal ablation, thermal stress and hyperthermia, mechanical perturbation, and histotripsy [184]. However, the first and foremost effect of HIFU is thermal ablation (by heating tumor tissue above approximately 55 °C), resulting in coagulative necrosis, combined with additional cavitation formation. The most interesting secondary effect is anti-tumor immunity induction [185,186].
The immunotherapeutic effect of HIFU has recently been investigated in many kinds of malignancies. Hu et al. confirmed HIFU promotes DC infiltration and activation in mice bearing colon adenocarcinoma and indicated that the mechanical components of this procedure may be successfully combined with other types of therapy [187]. Ran et al. showed that HIFU increases peripheral blood CD3+, CD4+ levels and the CD4+/CD8+ ratio, enhances CTL cytotoxicity against murine hepatocarcinoma, and inhibits tumor growth and progression in mice [188]. The impact on the CD4+/CD8+ ratio has been observed in the past by Rosberger et al. [189]. The activation of anti-tumor immunity promoted by HIFU can be partially explained by tumor debris “left-over” antigen immunogenicity, which was demonstrated by Zhang et al. in the murine hepatocellular carcinoma model [190]. Similar investigations have been conducted with other malignancies, such as melanoma, neuroblastoma, and pancreatic cancer [191,192,193,194,195,196]. Wu et al. researched tumor debris immunogenic properties in 23 patients with breast cancer. Using HIFU, they ablated primary tumors and evaluated the expression of tumor antigens and heat-shock protein 70 (HSP-70), while also pointing out the immunogenic potential of neoplastic debris [197].
Sonodynamic therapy (SDT) is another promising strategy concerning the usage of ultrasound. It is based on the application of sonosensitizers, which is followed by their activation with ultrasound. Activated particles then transfer the energy to oxygen accumulated in the TME, creating ROS which kill or damage tumor cells [198,199]. HIFU and the spectrum of ultrasound-based therapies in general are still very modern approaches used for enhancing immune response. Further investigation is required, especially concerning PC.

5.2. Cryotherapy

Cryoablation or cryotherapy performed either as a focal therapy, or as the whole-gland procedure, is an ablation technique using extremely low temperatures to induce both necrosis and apoptosis of tumor cells [200,201,202]. With the use of special cryoprobes, liquid nitrogen or argon, passing from high pressure to an atmospheric pressure revealing its cooling effect, is implemented inside a prostate gland. Although it may be used as a monotherapy, for this review, we will only focus on its immunomodulatory activity and its combination with immunotherapy.
Cryotherapy has great potential to enhance the immune response, due to its significant preservation of tumor antigens and cytokines, compared to other ablation techniques based on high temperatures and rather than hypothermia [203]. It is believed to leave the intracellular molecules of tumors intact and, through attracting the immune system using these factors, stimulate tumor-specific immunity. However, cryotherapy can prompt both immunostimulatory and immunosuppressive responses, which are strongly dependent on the type of induced cell death; studies suggest that necrosis, occurring mainly in the inner zone of the tissue, causes tumor cells to release danger-associated molecular patterns, which boost the immune response through the maturation of DCs, and consequently, T cell activation. However, apoptosis primarily occurring in the peripheral margin of the ablated organ leads to a lack of secretion of danger signals, therefore contributing to immunosuppression [204,205,206]. The cryoimmunological effect is further described by the term “abscopal effect”. This rare phenomenon refers to the systemic immunological impact a focal therapy has and primarily refers to the reduction in a metastasis preceded by a localized treatment in a different location [207]. This process was proved to be mediated by CD8+ T cells and correlated with a low level of CD4+CD5+ Tregs, as well as an increased level of IFN-ɣ [208,209].
Various investigations have been conducted on the theme of cryoimmunological therapy, both in murine models and in clinical trials. For instance, Gaitanis and Bassukas researched the impact of immunocryosurgery on basal cell carcinomas (BCC). Their study indicated that cryoablation combined with the TLR7 agonist, imiquimod, can be a very effective substitute for surgical treatment for BCC under 20 mm in diameter [210]. In another study, Lin et al. prospectively evaluated allogeneic NK cell immunotherapy combined with cryosurgery in renal cell carcinoma (RCC). They once again proved an additive effect of the two therapies [211]. The same group of researchers conducted similar investigations in patients with lung and hepatocellular cancers, with similarly favorable results [212,213].
So far clinical trials including the combination of cryosurgery and immunotherapy in patients with PC are rarely conducted. One of them is a therapy using granulocyte–macrophage colony-stimulating factor (GM-CSF), a cytokine regulating the function of granulocytes and macrophages, as well as promoting the survival of DCs [214,215]. These investigations revealed that GM-CSF administration enhances INF-ɣ secretion by T cells on the base of prior cryoablation procedures, as well as the fact that GM-CSF increases levels of prostate-specific and nonspecific antigens. Ross et al. examined cryosurgery combined with short term ADT and pembrolizumab, a PD-1 inhibitor, proving local disease control but questioning its potential for the management of systemic disease [216].

5.3. Photodynamic Therapy

PDT is an example of another targeted treatment option, that has already been used as an alternative to radical therapies, with the intention of reducing levels of side effects, while maintaining favorable oncological outcomes [217]. This focal therapy is based on the usage of a laser of a specific wavelength, which activates the photosensitizer (PS), administered systemically or locally, and therefore generates ROS resulting in necrosis of the tumor cells [218]. Depending on the qualities of photoagents, different effects can be achieved. Photothermal therapy (PTT) is a subtype of phototherapy that is different from PDT as it engages PS properties not to produce ROS but to execute a thermal effect through the conversion of absorbed laser light into heat [219].
As for PDT in PC, researchers point out the high efficacy and low level of adverse effects of vascular-targeted photodynamic therapy (VTP) in comparison to other therapies, while addressing the great need for long-term benefit evaluation in randomized clinical trials (RCT) [220]. Rastinehad et al. introduced the results of a clinical trial in which they used gold-silica nanoshells (AuroShells) to conduct PTT in 15 patients with PC. The study revealed high-profile feasibility of the procedure, and once again pointed out its low-rate adverse effects burden [221]. Another study by Azzouzi et al. compared padeliporfin VTP with an active surveillance strategy in a phase III RCT. They evaluated VTP as a safe and effective treatment for low-risk, localized PC, with a longer time to progression and a higher proportion of negative biopsy results in comparison to active surveillance [222]. On the other hand, a review of this investigation, initiated by the Oncologic Drugs Advisory Committee within the FDA, resulted in voting against approval of this therapeutic strategy in the United States [223]. There were many reasons for this, including, above all, an unfavorable benefit–risk profile of this therapy, postulated by the panelists [224]. Nevertheless, more clinical trials are required, evaluating different doses of various PS, varying laser wavelengths, and manipulating other parameters [223].
However, more and more papers recently have been turning their attention to the immunological aspects of PDT as it propagates inflammatory response, induces necrosis, and promotes recruitment of neutrophils and other immune cells [225,226,227,228,229]. Furthermore, PDT can promote immune cells and engage them to eradicate distant metastases [230]. Therefore, the term photoimmunotherapy (PIT) has been forged, and it may be described as a combination of immunogenic properties of PDT and immunotherapy treatment [231]. The immunological effect obtained by PDT is complex and multi-level. First of all, it directly affects immune cells through the recruitment of neutrophils, DC maturation, and macrophage activation, as well as the accumulation of CTLs and affecting them through regulation of NK cell migration [225,232]. Secretion of IL-1 α/β, IL-6, IL-8, IL-10, and IL-12 is boosted as is the release of a few secondary inflammatory mediators, including thromboxane and prostaglandins [233]. Furthermore, a few strategies concerning the combination of PDT with different immunotherapeutic strategies have been conducted, and their results are promising. Li et al. evaluated the synergistic effect of CTLA-4 antibodies and single-walled carbon nanotube-glycated chitosan complex (SWNT-GC), which acts primarily as a light absorber, in metastatic mammary tumors in mice. Local administration was then followed by PTT. The results showed that this strategy prolonged survival time, suppressed primary tumors, and inhibited metastases [234]. Huang et al. introduced a drug conjugate consisting of protoporphyrin IX and NLG919, a potent indoleamine-2,3-dioxygenase (IDO) inhibitor, which is applied to the cells through liposomal delivery (PpIX-NLG@Lipo). They showed its strong ability to generate ROS after the phototherapeutic procedure, as well as its potential to increase CD8+ T cell infiltration [235]. Kim et al. investigated the impact of PDT with a Ce6-embedded nanophotosensitizer (FIC-PDT) with ripasudil, a rho-kinase (ROCK) inhibitor, on the immune response in mice with uveal melanoma. Their research indicated that this combination demonstrates a vaccine-like function, realized by evoking immunogenic cell death and stimulating APCs, which leads to CD8+ T cell priming and their accumulation in the primary tumor, and, in further synergy with the anti-PD-L1 antibody, to metastasis inhibition [133].
Nagaya et al. presented the effects of near-infrared photoimmunotherapy (NIR-PIT) with a prostate-specific membrane antigen (PSMA) antibody in the PC cell line. The anti-PSMA antibody was conjugated to the light-absorbing agent, IR700DX. This antibody–PS conglomerate was observed to bind cell-specifically and to effectively kill PC cells after activation using NIR-PIT, with over two-thirds of the investigated tumors cured [236]. Research on the same topic was conducted by Watanabe et al., and it pointed to the possibility of using only fragments of anti-PSMA antibodies instead of the full antibodies, which may clinically translate to a more thorough penetration of the tumor milieu. Using smaller parts of antibodies should also shorten the time gap between the injection of the PS and NIR-PIT [237].

5.4. Irreversible Electroporation

IRE is the permeabilization of cell membranes with electrical pulses, which affect membranous electrochemical potentials, creating pores in a lipid bilayer [238]. IRE has been already used in PC management, both as a focal therapy, and as the whole gland ablation [239,240,241,242,243,244,245,246]. The procedure is based on needle electrodes, which are placed inside or nearby the targeted tissue. Then, short electrical pulses are delivered, which induces cell death through a non-thermal mechanism [247]. Despite its role in the immune response is still unexplored, IRE seems to have immunomodulatory activity. The most pivotal immunological effect of IRE is a decrease in Tregs in the TME; additionally, a decrease in MDSCs occurs as well [248].
The field of IRE–immunotherapy combinations in treating malignancies is still uninvestigated, although there are a few articles, especially on pancreatic cancer. Yang et al., for example, revealed a connection between IRE and tumor-associated immune evasion in a mice model of pancreatic ductal adenocarcinoma (PDAC). They indicated that IRE combined with the DC cancer vaccination increases the level of tumor-infiltrating cells including CD8+ T cells and granzyme B+ cells in PDAC [249]. Similar investigations have been conducted by Zhao et al. and by He et al. Both studies showed promising results from the combination of IRE and PD-1 inhibitors in mice with PDAC [250,251].
A study by Burbach et al. examined the combination of IRE and ICI in mice with PC. Focal treatment using IRE combined with ICI led to the expansion of tumor-specific CD8+ T cells in blood and the TME [252].

6. Radiation and Immune Therapy Combination

RT has been used as a management strategy both in PC and in many other malignancies for years. Its primary property, exploited for tumor treatment purposes, is the effect on double-strand DNA, leading to its breakdown, and thus resulting in cell death, majorly through senescence and slightly less frequently through mitotic catastrophe, apoptosis, and necrosis [253]. Traditionally, RT was considered to be a therapy of immunosuppressive qualities, therefore its combination with immunotherapy appeared to be irrational at first [254]. However, rapidly growing interest in the TME affected the way RT is perceived, as its relationship with the immune system is far more complex and ambiguous [255,256].

6.1. Immunomodulatory Effect of RT

An immune-stimulating effect of RT is generally achieved through induced cell death and modulating the composition of the TME [257]. One of the initial steps following tumor cell damage is the enhanced release of damage-associated molecules, such as calreticulin, adenosine triphosphate (ATP), GM-CSF, high-mobility group box 1 (HMGB1), and heat shock proteins (HSPs) [258,259]. Afterwards, these damage signals activate DCs and APCs, which takes place in lymph nodes and lead to priming naïve T cells as a consequence [259]. Additionally, one of the radiation effects is the release of other inflammatory molecules, such as chemokines (e.g., CXCL10 and CXCL16) and other cytokines, including IL-1β, TNF-α, and type 1 and 2 interferons, which further contribute to the increase in inflammation in the TME [260]. Finally, RT triggers upregulation of MHC I, the NKG2D ligand, Fas/CD95, and other co-stimulatory molecules, resulting in cell death and further antigen exposure [259,260].
Eckert et al. investigated the impact of RT on the immune system in 18 patients with localized PC. The study revealed the ambiguous effect of ionizing radiation in RT resulted in a decrease in absolute leukocyte and lymphocyte counts and an increase in Tregs and NK cells counts over eight weeks after radiation. However, during RT, an increase was observed in all immune cells counts excluding Tregs. Importantly, the percentage of CD8+ T cells showed its peak early during RT [261]. Nevertheless, Harris et al. researched a combination of RT and immunotherapy in a transgenic murine model and observed that the anti-tumor immune response occurred when immune therapy was administered 3 to 5 weeks after RT [20]. This further suggests the existence of a certain type of therapeutic time window, in which the immunostimulatory properties of RT are emphasized, and the immunosuppressive component is partially inhibited. Nickols et al. researched the impact of stereotactic body radiotherapy (SBRT) on immunological homeostasis in a clinical trial evaluating the resected prostate specimens of 16 patients. While prostates without SBRT were mainly lymphoid diverse, specimens after SBRT were immunologically dominated by myeloid cells [262]. Keam et al. proved in their 24 patient clinical trial that high dose-rate brachytherapy (HDRBT) has substantial potential in enhancing inflammation in the prostate. In response to HDRBT, an increase in CD4+ T cells, macrophages, and DCs counts was observed. Moreover, they evaluated the tumor inflammation signature (TIS) and concluded that 80% of immunologically “cold” tumors were converted to “intermediate” or “hot” types [27].
Interestingly, RT is another management strategy with proven abscopal effect, hence resulting in the regression of metastases, probably due to the outburst of tumor-associated antigens. This extremely rare effect is observed more often when RT is combined with immune therapy, particularly with checkpoint inhibitors [258]. Dudzinski et al. studied the combination of anti-PD-1 or anti-PD-L1 and radiation in mice, and they not only observed an increase in median survival in comparison to the drug alone (70% longer for anti-PD-1 and 130% for anti-PD-L1) but also detected the abscopal effect—a regression of unirradiated distant metastases [263].

6.2. Radioimmunotherapy in Murine Models

In the research concerning the effects of RT and immunotherapy combination in mice, there have been a few distinguishing articles, including the paper from Wada et al. They assessed the efficacy of this therapy (the immunotherapeutic component was GM-CSF) using an autochthonous model of PC. Improved OS and increase in the effector-to-regulatory TILs ratio, as well as treatment effect in both the primary tumor and metastases, were observed [264]. Another investigation by Philippou et al. assayed the combination of anti-PD-L1 and RT and its impact on the TME in PC. They observed macrophage and DC counts increase, as well as an upregulation of PD-1/PD-L1 in both arms of the study 7 days after RT. Radiation was observed to delay tumor growth and affect the TME immunological composition. However, PD-L1 inhibition administered in one of the arms did not affect tumor growth delay when compared to monotherapy [265]. Table 1 presents ongoing trials evaluating different combinations of RT and immunotherapy in PC management.

6.3. Radioimmunotherapy in Prostate Cancer

The efficiency of radioimmunotherapy in patients with PC has been willingly explored in clinical trials for the last 10 years. Slovin et al. assessed the combination of anti-CTLA-4 antibody, ipilimumab, with external beam radiotherapy (EBRT) in comparison to the drug alone. This phase I/II study on 50 patients evaluated adverse effects, defining them as manageable and indicating anti-tumor activity [266]. In another investigation, a phase III trial concerning ipilimumab versus placebo after radiotherapy in patients with mCRPC that progressed after docetaxel chemotherapy was conducted by Kwon et al. No notable difference in OS was found, although ipilimumab use was associated with a decrease in PSA levels and an increase in PFS. Additionally, an OS increase was observed in the ipilimumab subgroup without visceral metastases, with non-raised or mildly raised alkaline phosphatase and without anemia. Accordingly, the authors suggested that a specific constellation of prognostic features could potentially enhance the clinical outcomes of radioimmunotherapy [267]. The final analysis of this phase III trial revealed that OS was two to three times higher at 3 years and beyond in favor of the radiotherapy and ipilimumab combination [267]. Different clinical trials assessing nivolumab and brachytherapy or EBRT, as well as sipuleucel-T and EBRT combinations indicated that these therapies are safe and well-tolerated. The immunogenic effect and anti-tumor activity of radiation with nivolumab were observed, while radiation with sipuleucel-T showed no particular increase in the immune response [268,269]. Another phase II trial assessed the combination of sipuleucel-T and a radioisotope, radium-223, in patients with mCRPC. Despite paradoxically decreasing the immune response in the combination arm, PSA levels were decreased, and PFS and OS were longer [270]. A case report by Han et al. presents a significant clinical response to the pembrolizumab and radiation combination in a patient heavily treated for mCRPC with rectal involvement. After radiation and six cycles of the drug, the PSA was undetectable, the prostate mass was decreased, and the rectal invasion was imperceptible in imaging studies [271].

7. Overview of the Immunomodulatory Effects of Focal Therapies and RT

Focal therapies and RT boost the immune response in multiple ways. As previously mentioned, it is difficult to make an accurate comparison of the immunomodulatory effects due to miscellaneous reasons, including the complexity of the immune system, a variety of possible immunological outcomes, and lack of clinical trials assessing the differences between therapies. Moreover, it is worth mentioning that most studies do not distinguish an additive effect of a therapy from a synergistic effect, which concerns studies on cytokines expression, in particular, but there are articles that correctly incorporate both terms [272,273,274,275,276]. In the case of studying the immunomodulatory effects of focal therapies, an additive effect may be defined if the level or value of a certain immune property is either equal or lower in the combinational approach than the sum of the values of the effect in the monotherapeutic approach (1 + 1 ≤ 2). A synergistic effect occurs when the level of analyzed immune effect is higher in the combinational approach than the sum of the values in monotherapies alone (1 + 1 ≥ 2) [277,278]. This nuance may have clinical implications in future studies and should be carefully analyzed in multi-arm clinical trials assessing the immune effect of RT and focal therapies.
Table 2 presents a thorough overview of the immunomodulatory impact of different local treatment strategies on the TME.

8. Conclusions

Immunotherapy for PC has been thoroughly explored in recent years. Despite the initial success of sipuleucel-T, it seems that immunotherapy shows lower efficacy in PC in comparison to other malignancies. Further phase I/II clinical trials investigating radioimmunotherapy and combinations of focal and immune therapies are highly desirable. The first studies concerning the immunomodulatory properties of RT and focal therapies cautiously showed promising results, although a well-planned randomized clinical trial, addressing both immunological and clinical outcomes, would be necessary to accurately assess the usefulness of immunotherapy and its combinations in PC management. A meticulously designed study such as this could plausibly prove the synergistic effect provided by the combination of therapies, which would hopefully create new clinical perspectives for focal treatment options.

Author Contributions

Conceptualization, J.K. (Jakub Karwacki), A.K., and B.M.; validation, J.K. (Jakub Karwacki), A.K., W.S., J.K. (Julita Kulbacka), K.K., B.M., and J.S.; formal analysis, J.K., A.K., and N.S.; data curation, J.K. (Jakub Karwacki), W.K., and W.S.; writing—original draft preparation, J.K. (Jakub Karwacki), A.K., and B.M.; writing—review and editing, J.K. (Jakub Karwacki); visualization, W.S. and N.S.; supervision, B.M., A.K., T.S., J.K. (Julita Kulbacka), and J.S.; project administration, B.M.; funding acquisition, B.M. All authors have read and agreed to the published version of the manuscript.

Funding

The paper was partially funded by the Department of Molecular and Cellular Biology, no. SUB.D260.22.016 and partially funded by the University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, no. SUBZ.C090.22.057.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

Figure 1 was partly generated using Servier Medical Art (http://smart.servier.com/, accessed on 11 July 2022), provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. He, L.; Fang, H.; Chen, C.; Wu, Y.; Wang, Y.; Ge, H.; Wang, L.; Wan, Y.; He, H. Metastatic castration-resistant prostate cancer: Academic insights and perspectives through bibliometric analysis. Medicine 2020, 99, e19760. [Google Scholar] [CrossRef] [PubMed]
  3. Ahdoot, M.; Lebastchi, A.H.; Turkbey, B.; Wood, B.; Pinto, P.A. Contemporary treatments in prostate cancer focal therapy. Curr. Opin. Oncol. 2019, 31, 200–206. [Google Scholar] [CrossRef]
  4. May, K.F.; Gulley, J.L.; Drake, C.G.; Dranoff, G.; Kantoff, P.W. Prostate Cancer Immunotherapy. Clin. Cancer Res. 2011, 17, 5233–5238. [Google Scholar] [CrossRef]
  5. Bansal, D.; Reimers, M.; Knoche, E.; Pachynski, R. Immunotherapy and Immunotherapy Combinations in Metastatic Castration-Resistant Prostate Cancer. Cancers 2021, 13, 334. [Google Scholar] [CrossRef]
  6. Huang, L.; LaBonte, M.J.; Craig, S.G.; Finn, S.P.; Allott, E.H. Inflammation and Prostate Cancer: A Multidisciplinary Approach to Identifying Opportunities for Treatment and Prevention. Cancers 2022, 14, 1367. [Google Scholar] [CrossRef]
  7. Vitkin, N.; Nersesian, S.; Siemens, D.R.; Koti, M. The Tumor Immune Contexture of Prostate Cancer. Front. Immunol. 2019, 10, 603. [Google Scholar] [CrossRef] [PubMed]
  8. Carvalho, H.d.A.; Villar, R.C. Radiotherapy and immune response: The systemic effects of a local treatment. Clinics 2018, 73 (Suppl. S1), e557s. [Google Scholar] [CrossRef] [PubMed]
  9. Borges, R.C.; Tourinho-Barbosa, R.R.; de la Rosette, J. Tumour microenvironment and focal therapy for prostate cancer. Curr. Opin. Urol. 2022, 32, 248–253. [Google Scholar] [CrossRef] [PubMed]
  10. Chavez, M.; Silvestrini, M.T.; Ingham, E.S.; Fite, B.Z.; Mahakian, L.M.; Tam, S.M.; Ilovitsh, A.; Monjazeb, A.M.; Murphy, W.J.; Hubbard, N.E.; et al. Distinct immune signatures in directly treated and distant tumors result from TLR adjuvants and focal ablation. Theranostics 2018, 8, 3611–3628. [Google Scholar] [CrossRef]
  11. Dai, J.; Lu, Y.; Roca, H.; Keller, J.M.; Zhang, J.; McCauley, L.K.; Keller, E.T. Immune mediators in the tumor microenvironment of prostate cancer. Chin. J. Cancer 2017, 36, 29. [Google Scholar] [CrossRef] [PubMed]
  12. Giraldo, N.A.; Sanchez-Salas, R.; Peske, J.D.; Vano, Y.A.; Becht, E.; Petitprez, F.; Validire, P.; Ingels, A.; Cathelineau, X.; Fridman, W.H.; et al. The clinical role of the TME in solid cancer. Br. J. Cancer 2018, 120, 45–53. [Google Scholar] [CrossRef] [PubMed]
  13. Donkor, M.K.; Sarkar, A.; Savage, P.A.; Franklin, R.A.; Johnson, L.K.; Jungbluth, A.A.; Allison, J.P.; Li, M.O. T Cell Surveillance of Oncogene-Induced Prostate Cancer Is Impeded by T Cell-Derived TGF-β1 Cytokine. Immunity 2011, 35, 123–134. [Google Scholar] [CrossRef]
  14. Kwon, J.T.W.; Bryant, R.J.; Parkes, E.E. The tumor microenvironment and immune responses in prostate cancer patients. Endocr. -Relat. Cancer 2021, 28, T95–T107. [Google Scholar] [CrossRef] [PubMed]
  15. Chen, D.S.; Mellman, I. Oncology Meets Immunology: The Cancer-Immunity Cycle. Immunity 2013, 39, 1–10. [Google Scholar] [CrossRef]
  16. Angell, H.; Galon, J. From the immune contexture to the Immunoscore: The role of prognostic and predictive immune markers in cancer. Curr. Opin. Immunol. 2013, 25, 261–267. [Google Scholar] [CrossRef] [PubMed]
  17. Gao, G.; Wang, Z.; Qu, X.; Zhang, Z. Prognostic value of tumor-infiltrating lymphocytes in patients with triple-negative breast cancer: A systematic review and meta-analysis. BMC Cancer 2020, 20, 179. [Google Scholar] [CrossRef]
  18. Ropponen, K.M.; Eskelinen, M.J.; Lipponen, P.K.; Alhava, E.; Kosma, V.-M. Prognostic value of tumour-infiltrating lymphocytes (TILs) in colorectal cancer. J. Pathol. 1997, 182, 318–324. [Google Scholar] [CrossRef]
  19. Fu, Q.; Chen, N.; Ge, C.; Li, R.; Li, Z.; Zeng, B.; Li, C.; Wang, Y.; Xue, Y.; Song, X.; et al. Prognostic value of tumor-infiltrating lymphocytes in melanoma: A systematic review and meta-analysis. OncoImmunology 2019, 8, e1593806. [Google Scholar] [CrossRef] [PubMed]
  20. Harris, T.J.; Hipkiss, E.L.; Borzillary, S.; Wada, S.; Grosso, J.F.; Yen, H.-R.; Getnet, D.; Bruno, T.C.; Goldberg, M.V.; Pardoll, D.M.; et al. Radiotherapy augments the immune response to prostate cancer in a time-dependent manner. Prostate 2008, 68, 1319–1329. [Google Scholar] [CrossRef] [Green Version]
  21. Li, J.; Wang, J.; Chen, R.; Bai, Y.; Lu, X. The prognostic value of tumor-infiltrating T lymphocytes in ovarian cancer. Oncotarget 2017, 8, 15621–15631. [Google Scholar] [CrossRef]
  22. Ding, W.; Xu, X.; Qian, Y.; Xue, W.; Wang, Y.; Du, J.; Jin, L.; Tan, Y. Prognostic value of tumor-infiltrating lymphocytes in hepatocellular carcinoma. Medicine 2018, 97, e13301. [Google Scholar] [CrossRef]
  23. De Bono, J.S.; Guo, C.; Gurel, B.; De Marzo, A.M.; Sfanos, K.S.; Mani, R.S.; Gil, J.; Drake, C.G.; Alimonti, A. Prostate carcinogenesis: Inflammatory storms. Nat. Cancer 2020, 20, 455–469. [Google Scholar] [CrossRef] [PubMed]
  24. Yang, Y.; Attwood, K.; Bshara, W.; Mohler, J.L.; Guru, K.; Xu, B.; Kalinski, P.; Chatta, G. High intratumoral CD8+ T-cell infiltration is associated with improved survival in prostate cancer patients undergoing radical prostatectomy. Prostate 2020, 81, 20–28. [Google Scholar] [CrossRef]
  25. Vicier, C.; Werner, L.; Huang, Y.; Hamid, A.; Evan, C.; Loda, M.; Sweeney, C. Immune infiltrate with CD8 low or PDL1 high associated with metastatic prostate cancer after radical prostatectomy (RP). J. Clin. Oncol. 2019, 37, 86. [Google Scholar] [CrossRef]
  26. Ness, N.; Andersen, S.; Valkov, A.; Nordby, Y.; Donnem, T.; Al-Saad, S.; Busund, L.-T.; Bremnes, R.M.; Richardsen, E. Infiltration of CD8+ lymphocytes is an independent prognostic factor of biochemical failure-free survival in prostate cancer. Prostate 2014, 74, 1452–1461. [Google Scholar] [CrossRef] [PubMed]
  27. Keam, S.P.; Halse, H.; Nguyen, T.; Wang, M.; Losio, N.V.K.; Mitchell, C.; Caramia, F.; Byrne, D.J.; Haupt, S.; Ryland, G.; et al. High dose-rate brachytherapy of localized prostate cancer converts tumors from cold to hot. J. Immunother. Cancer 2020, 8, e000792. [Google Scholar] [CrossRef]
  28. Hammerstrom, A.E.; Cauley, D.H.; Atkinson, B.J.; Sharma, P. Cancer Immunotherapy: Sipuleucel-T and Beyond. Pharmacother. J. Hum. Pharmacol. Drug Ther. 2011, 31, 813–828. [Google Scholar] [CrossRef]
  29. Melichar, B.; Nash, M.A.; Lenzi, R.; Platsoucas, C.D.; Freedman, R.S. Expression of costimulatory molecules CD80 and CD86 and their receptors CD28, CTLA-4 on malignant ascites CD3+ tumour-infiltrating lymphocytes (TIL) from patients with ovarian and other types of peritoneal carcinomatosis. Clin. Exp. Immunol. 2000, 119, 19–27. [Google Scholar] [CrossRef]
  30. Nakajima, A.; Watanabe, N.; Yoshino, S.; Yagita, H.; Okumura, K.; Azuma, M. Requirement of CD28-CD86 co-stimulation in the interaction between antigen-primed T helper type 2 and B cells. Int. Immunol. 1997, 9, 637–644. [Google Scholar] [CrossRef] [Green Version]
  31. Takahashi, T.; Tagami, T.; Yamazaki, S.; Uede, T.; Shimizu, J.; Sakaguchi, N.; Mak, T.W.; Sakaguchi, S. Immunologic Self-Tolerance Maintained by Cd25+Cd4+Regulatory T Cells Constitutively Expressing Cytotoxic T Lymphocyte–Associated Antigen. J. Exp. Med. 2000, 192, 303–310. [Google Scholar] [CrossRef]
  32. Alegre, M.-L.; Frauwirth, K.A.; Thompson, C.B. T-cell regulation by CD28 and CTLA-4. Nat. Rev. Immunol. 2001, 1, 220–228. [Google Scholar] [CrossRef]
  33. Rudd, C.E.; Taylor, A.; Schneider, H. CD28 and CTLA-4 coreceptor expression and signal transduction. Immunol. Rev. 2009, 229, 12–26. [Google Scholar] [CrossRef]
  34. Sansom, D. CD28, CTLA-4 and their ligands: Who does what and to whom? Immunology 2000, 101, 169–177. [Google Scholar] [CrossRef]
  35. Krummel, M.F.; Allison, J. CTLA-4 engagement inhibits IL-2 accumulation and cell cycle progression upon activation of resting T cells. J. Exp. Med. 1996, 183, 2533–2540. [Google Scholar] [CrossRef]
  36. Belani, R.; Weiner, G.J. Expression of both B7-1 and CD28 contributes to the IL-2 responsiveness of CTLL-2 cells. Immunology 1996, 87, 271–274. [Google Scholar] [CrossRef]
  37. Loke, P.; Allison, J.P. PD-L1 and PD-L2 are differentially regulated by Th1 and Th2 cells. Proc. Natl. Acad. Sci. USA 2003, 100, 5336–5341. [Google Scholar] [CrossRef] [PubMed]
  38. Latchman, Y.; Wood, C.R.; Chernova, T.; Chaudhary, D.; Borde, M.; Chernova, I.; Iwai, Y.; Long, A.J.; Brown, J.A.; Nunes, R.; et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat. Immunol. 2001, 2, 261–268. [Google Scholar] [CrossRef]
  39. Wang, L.; Pino-lagos, K.; De Vries, V.C.; Guleria, I.; Sayegh, M.H.; Noelle, R.J. Programmed death 1 ligand signaling regulates the generation of adaptive Foxp3+CD4+ regulatory T cells. Proc. Natl. Acad. Sci. USA 2008, 105, 9331–9336. [Google Scholar]
  40. Brown, J.A.; Dorfman, D.M.; Ma, F.-R.; Sullivan, E.L.; Munoz, O.; Wood, C.R.; Greenfield, E.A.; Freeman, G.J. Blockade of Programmed Death-1 Ligands on Dendritic Cells Enhances T Cell Activation and Cytokine Production. J. Immunol. 2003, 170, 1257–1266. [Google Scholar] [CrossRef]
  41. Blank, C.; Gajewski, T.F.; Mackensen, A. Interaction of PD-L1 on tumor cells with PD-1 on tumor-specific T cells as a mechanism of immune evasion: Implications for tumor immunotherapy. Cancer Immunol. Immunother. 2004, 54, 307–314. [Google Scholar] [CrossRef] [PubMed]
  42. Jafari, S.; Molavi, O.; Kahroba, H.; Hejazi, M.S.; Maleki-Dizaji, N.; Barghi, S.; Kiaie, S.H.; Jadidi-Niaragh, F. Clinical application of immune checkpoints in targeted immunotherapy of prostate cancer. Cell. Mol. Life Sci. 2020, 77, 3693–3710. [Google Scholar] [CrossRef]
  43. Vinay, D.S.; Ryan, E.P.; Pawelec, G.; Talib, W.H.; Stagg, J.; Elkord, E.; Lichtor, T.; Decker, W.K.; Whelan, R.L.; Kumara, H.M.C.S.; et al. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin. Cancer Biol. 2015, 35, S185–S198. [Google Scholar] [CrossRef]
  44. Jing, X.; Yang, F.; Shao, C.; Wei, K.; Xie, M.; Shen, H.; Shu, Y. Role of hypoxia in cancer therapy by regulating the tumor microenvironment. Mol. Cancer 2019, 18, 157. [Google Scholar] [CrossRef]
  45. Wang, B.; Zhao, Q.; Zhang, Y.; Liu, Z.; Zheng, Z.; Liu, S.; Meng, L.; Xin, Y.; Jiang, X. Targeting hypoxia in the tumor microenvironment: A potential strategy to improve cancer immunotherapy. J. Exp. Clin. Cancer Res. 2021, 40, 24. [Google Scholar] [CrossRef]
  46. Chang, C.-H.; Qiu, J.; O’Sullivan, D.; Buck, M.D.; Noguchi, T.; Curtis, J.D.; Chen, Q.; Gindin, M.; Gubin, M.M.; van der Windt, G.J.W.; et al. Metabolic Competition in the Tumor Microenvironment Is a Driver of Cancer Progression. Cell 2015, 162, 1229–1241. [Google Scholar] [CrossRef] [PubMed]
  47. Sasidharan Nair, V.; Elkord, E. Immune checkpoint inhibitors in cancer therapy: A focus on T-regulatory cells. Immunol. Cell Biol. 2018, 96, 21–33. [Google Scholar] [CrossRef]
  48. Wolf, A.M.; Wolf, D.; Steurer, M.; Gastl, G.; Gunsilius, E.; Grubeck-Loebenstein, B. Increase of regulatory T cells in the peripheral blood of cancer patients. Clin. Cancer Res. 2003, 9, 606–612. [Google Scholar]
  49. Dannull, J.; Su, Z.; Rizzieri, D.; Yang, B.K.; Coleman, D.; Yancey, D.; Zhang, A.; Dahm, P.; Chao, N.; Gilboa, E.; et al. Enhancement of vaccine-mediated antitumor immunity in cancer patients after depletion of regulatory T cells. J. Clin. Investig. 2005, 115, 3623–3633. [Google Scholar] [CrossRef]
  50. Miller, A.M.; Lundberg, K.; Özenci, V.; Banham, A.H.; Hellström, M.; Egevad, L.; Pisa, P. CD4+CD25high T Cells Are Enriched in the Tumor and Peripheral Blood of Prostate Cancer Patients. J. Immunol. 2006, 177, 7398–7405. [Google Scholar] [CrossRef]
  51. Karpisheh, V.; Mousavi, S.M.; Sheykholeslami, P.N.; Fathi, M.; Saray, M.M.; Aghebati-Maleki, L.; Jafari, R.; Zolbanin, N.M.; Jadidi-Niaragh, F. The role of regulatory T cells in the pathogenesis and treatment of prostate cancer. Life Sci. 2021, 284, 119132. [Google Scholar] [CrossRef] [PubMed]
  52. Houghton, A.N.; Guevara-Patiño, J.A. Immune recognition of self in immunity against cancer. J. Clin. Investig. 2004, 114, 468–471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Touloukian, E.C.; Leitner, W.W.; Robbins, P.F.; Li, Y.F.; Kang, X.; Lapointe, R.; Hwu, P.; A Rosenberg, S.; Restifo, N.P. Expression of a “self-”antigen by human tumor cells enhances tumor antigen-specific CD4(+) T-cell function. Cancer Res. 2002, 62, 5144–5147. [Google Scholar] [PubMed]
  54. Bunt, S.K.; Yang, L.; Sinha, P.; Clements, V.K.; Leips, J.; Ostrand-Rosenberg, S. Reduced Inflammation in the Tumor Microenvironment Delays the Accumulation of Myeloid-Derived Suppressor Cells and Limits Tumor Progression. Cancer Res. 2007, 67, 10019–10026. [Google Scholar] [CrossRef] [PubMed]
  55. Choi, J.-N.; Sun, E.G.; Cho, S.-H. IL-12 Enhances Immune Response by Modulation of Myeloid Derived Suppressor Cells in Tumor Microenvironment. Chonnam. Med. J. 2019, 55, 31–39. [Google Scholar] [CrossRef] [PubMed]
  56. Li, H.; Han, Y.; Guo, Q.; Zhang, M.; Cao, X. Cancer-expanded myeloid-derived suppressor cells induce anergy of NK cells through membrane-bound TGF-β1. J. Immunol. 2009, 182, 240–249. [Google Scholar] [CrossRef]
  57. Serafini, P.; Mgebroff, S.; Noonan, K.; Borrello, I. Myeloid-Derived Suppressor Cells Promote Cross-Tolerance in B-Cell Lymphoma by Expanding Regulatory T Cells. Cancer Res. 2008, 68, 5439–5449. [Google Scholar] [CrossRef]
  58. Hoechst, B.; Ormandy, L.A.; Ballmaier, M.; Lehner, F.; Krüger, C.; Manns, M.P.; Greten, T.F.; Korangy, F. A New Population of Myeloid-Derived Suppressor Cells in Hepatocellular Carcinoma Patients Induces CD4+CD25+Foxp3+ T Cells. Gastroenterology 2008, 135, 234–243. [Google Scholar] [CrossRef]
  59. Srivastava, M.K.; Sinha, P.; Clements, V.K.; Rodriguez, P.; Ostrand-Rosenberg, S. Myeloid-Derived Suppressor Cells Inhibit T-Cell Activation by Depleting Cystine and Cysteine. Cancer Res. 2010, 70, 68–77. [Google Scholar] [CrossRef] [PubMed]
  60. Sanaei, M.J.; Salimzadeh, L.; Bagheri, N. Crosstalk between myeloid-derived suppressor cells and the immune system in prostate cancer. J. Leukoc. Biol. 2019, 107, 43–56. [Google Scholar] [CrossRef]
  61. Chi, N.; Tan, Z.; Ma, K.; Bao, L.; Yun, Z. Increased circulating myeloid-derived suppressor cells correlate with cancer stages, interleukin-8 and -6 in prostate cancer. Int. J. Clin. Exp. Med. 2014, 7, 3181–3192. [Google Scholar]
  62. Bosas, P.; Zaleskis, G.; Dabkevičiene, D.; Dobrovolskiene, N.; Mlynska, A.; Tikuišis, R.; Ulys, A.; Pašukoniene, V.; Jarmalaitė, S.; Jankevičius, F. Immunophenotype Rearrangement in Response to Tumor Excision May Be Related to the Risk of Biochemical Recurrence in Prostate Cancer Patients. J. Clin. Med. 2021, 10, 3709. [Google Scholar] [CrossRef] [PubMed]
  63. Siemińska, I.; Rychlicka-Buniowska, E.; Jaszczyński, J.; Palaczyński, M.; Bukowska-Strakova, K.; Ryś, J.; Dumański, J.; Siedlar, M.; Baran, J. The level of myeloid derived-suppressor cells in peripheral blood of patients with prostate cancerafter various types of therapy. Pol. J. Pathol. 2020, 71, 46–54. [Google Scholar] [CrossRef]
  64. Murugaiyan, G.; Martin, S.; Saha, B. Levels of CD40 expression on dendritic cells dictate tumour growth or regression. Clin. Exp. Immunol. 2007, 149, 194–202. [Google Scholar] [CrossRef] [PubMed]
  65. Guery, J.-C.; Adorini, L. Dendritic cells are the most efficient in presenting endogenous naturally processed self-epitopes to class II-restricted T cells. J. Immunol. 1995, 154, 536–544. [Google Scholar]
  66. Wykes, M.; MacPherson, G. Dendritic cell-B-cell interaction: Dendritic cells provide B cells with CD40-independent proliferation signals and CD40-dependent survival signals. Immunology 2000, 100, 1–3. [Google Scholar] [CrossRef]
  67. Wesa, A.; Galy, A. Increased production of pro-inflammatory cytokines and enhanced T cell responses after activation of human dendritic cells with IL-1 and CD40 ligand. BMC Immunol. 2002, 3, 14. [Google Scholar] [CrossRef]
  68. Hernandez, M.G.H.; Shen, L.; Rock, K.L. CD40-CD40 Ligand Interaction between Dendritic Cells and CD8+T Cells Is Needed to Stimulate Maximal T Cell Responses in the Absence of CD4+T Cell Help. J. Immunol. 2007, 178, 2844–2852. [Google Scholar] [CrossRef]
  69. Hill, K.S.; Errington, F.; Steele, L.P.; Merrick, A.; Morgan, R.; Selby, P.J.; Georgopoulos, N.T.; O’Donnell, D.M.; Melcher, A.A. OK432-Activated Human Dendritic Cells Kill Tumor Cells via CD40/CD40 Ligand Interactions. J. Immunol. 2008, 181, 3108–3115. [Google Scholar] [CrossRef]
  70. Wykes, M.; Pombo, A.; Jenkins, C.; MacPherson, G.G. Dendritic cells interact directly with naive B lymphocytes to transfer antigen and initiate class switching in a primary T-dependent response. J. Immunol. 1998, 161, 1313–1319. [Google Scholar]
  71. Kelsall, B.L.; Stüber, E.; Neurath, M.; Strober, W. Interleukin-12 Production by Dendritic Cells. Ann. N. Y. Acad. Sci. 1996, 795, 116–126. [Google Scholar] [CrossRef] [PubMed]
  72. Ma, D.Y.; Clark, E.A. The role of CD40 and CD154/CD40L in dendritic cells. Semin. Immunol. 2009, 21, 265–272. [Google Scholar] [CrossRef]
  73. Dewaalmalefyt, R.; Roncarolo, M.G.; Spits, H.; Devries, J. Interleukin-10. Curr. Opin. Immunol. 1992, 4, 314–320. [Google Scholar] [CrossRef]
  74. Solinas, G.; Germano, G.; Mantovani, A.; Allavena, P. Tumor-associated macrophages (TAM) as major players of the cancer-related inflammation. J. Leukoc. Biol. 2009, 86, 1065–1073. [Google Scholar] [CrossRef]
  75. Schoppmann, S.F.; Birner, P.; Stöckl, J.; Kalt, R.; Ullrich, R.; Caucig, C.; Kriehuber, E.; Nagy, K.; Alitalo, K.; Kerjaschki, D. Tumor-Associated Macrophages Express Lymphatic Endothelial Growth Factors and Are Related to Peritumoral Lymphangiogenesis. Am. J. Pathol. 2002, 161, 947–956. [Google Scholar] [CrossRef] [Green Version]
  76. Larionova, I.; Tuguzbaeva, G.; Ponomaryova, A.; Stakheyeva, M.; Cherdyntseva, N.; Pavlov, V.; Choinzonov, E.; Kzhyshkowska, J. Tumor-Associated Macrophages in Human Breast, Colorectal, Lung, Ovarian and Prostate Cancers. Front. Oncol. 2020, 10, 566511. [Google Scholar] [CrossRef] [PubMed]
  77. Yuri, P.; Shigemura, K.; Kitagawa, K.; Hadibrata, E.; Risan, M.; Zulfiqqar, A.; Soeroharjo, I.; Hendri, A.Z.; Danarto, R.; Ishii, A.; et al. Increased tumor-associated macrophages in the prostate cancer microenvironment predicted patients’ survival and responses to androgen deprivation therapies in Indonesian patients cohort. Prostate Int. 2020, 8, 62–69. [Google Scholar] [CrossRef] [PubMed]
  78. Gollapudi, K.; Galet, C.; Grogan, T.; Zhang, H.; Said, J.W.; Huang, J.; Elashoff, D.; Freedland, S.J.; Rettig, M.; Aronson, W.J. Association between tumor-associated macrophage infiltration, high grade prostate cancer, and biochemical recurrence after radical prostatectomy. Am. J. Cancer Res. 2013, 3, 523–529. [Google Scholar]
  79. Stultz, J.; Fong, L. How to turn up the heat on the cold immune microenvironment of metastatic prostate cancer. Prostate Cancer Prostatic Dis. 2021, 24, 697–717. [Google Scholar] [CrossRef] [PubMed]
  80. Mizutani, K.; Sud, S.; McGregor, N.A.; Martinovski, G.; Rice, B.T.; Craig, M.J.; Varsos, Z.S.; Roca, H.; Pienta, K.J. The Chemokine CCL2 Increases Prostate Tumor Growth and Bone Metastasis through Macrophage and Osteoclast Recruitment. Neoplasia 2009, 11, 1235–1242. [Google Scholar] [CrossRef]
  81. Ogiya, R.; Niikura, N.; Kumaki, N.; Yasojima, H.; Iwasa, T.; Kanbayashi, C.; Oshitanai, R.; Tsuneizumi, M.; Watanabe, K.-I.; Matsui, A.; et al. Comparison of immune microenvironments between primary tumors and brain metastases in patients with breast cancer. Oncotarget 2017, 8, 103671–103681. [Google Scholar] [CrossRef]
  82. Shibutani, M.; Maeda, K.; Nagahara, H.; Fukuoka, T.; Matsutani, S.; Kashiwagi, S.; Tanaka, H.; Hirakawa, K.; Ohira, M. A comparison of the local immune status between the primary and metastatic tumor in colorectal cancer: A retrospective study. BMC Cancer 2018, 18, 371. [Google Scholar] [CrossRef] [PubMed]
  83. Lee, H.; Na, K.J.; Choi, H. Differences in Tumor Immune Microenvironment in Metastatic Sites of Breast Cancer. Front. Oncol. 2021, 11, 649004. [Google Scholar] [CrossRef]
  84. Ye, Z.H.; Long, H.; Zhao, Z.R. Different in Situ Immune Patterns between Primary Tumor and Lymph Node in Non-Small-Cell Lung Cancer: Potential Impact on Neoadjuvant Immunotherapy. J. Immunol. Res. 2022, 2022, 8513747. [Google Scholar] [CrossRef]
  85. Szekely, B.; Bossuyt, V.; Li, X.; Wali, V.; Patwardhan, G.; Frederick, C.; Silber, A.; Park, T.; Harigopal, M.; Pelekanou, V.; et al. Immunological differences between primary and metastatic breast cancer. Ann. Oncol. 2018, 29, 2232–2239. [Google Scholar] [CrossRef] [PubMed]
  86. Cacho-Díaz, B.; García-Botello, D.R.; Wegman-Ostrosky, T.; Reyes-Soto, G.; Ortiz-Sánchez, E.; Herrera-Montalvo, L.A. Tumor microenvironment differences between primary tumor and brain metastases. J. Transl. Med. 2020, 18, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Ikarashi, D.; Okimoto, T.; Shukuya, T.; Onagi, H.; Hayashi, T.; Sinicropi-Yao, S.L.; Amann, J.M.; Nakatsura, T.; Kitano, S.; Carbone, D.P. Comparison of Tumor Microenvironments Between Primary Tumors and Brain Metastases in Patients With NSCLC. JTO Clin. Res. Rep. 2021, 2, 100230. [Google Scholar] [CrossRef]
  88. Woo, J.R.; Liss, A.M.; Muldong, M.T.; Palazzi, K.; Strasner, A.; Ammirante, M.; Varki, N.; Shabaik, A.; Howell, S.; Kane, C.J.; et al. Tumor infiltrating B-cells are increased in prostate cancer tissue. J. Transl. Med. 2014, 12, 30. [Google Scholar] [CrossRef] [PubMed]
  89. Ammirante, M.; Luo, J.-L.; Grivennikov, S.; Nedospasov, S.; Karin, M. B-cell-derived lymphotoxin promotes castration-resistant prostate cancer. Nature 2010, 464, 302–305. [Google Scholar] [CrossRef]
  90. Adekoya, T.O.; Richardson, R.M. Cytokines and Chemokines as Mediators of Prostate Cancer Metastasis. Int. J. Mol. Sci. 2020, 21, 4449. [Google Scholar] [CrossRef] [PubMed]
  91. Konkel, J.E.; Zhang, D.; Zanvit, P.; Chia, C.; Zangarle-Murray, T.; Jin, W.; Wang, S.; Chen, W. Transforming Growth Factor-β Signaling in Regulatory T Cells Controls T Helper-17 Cells and Tissue-Specific Immune Responses. Immunity 2017, 46, 660–674. [Google Scholar] [CrossRef]
  92. Massagué, J. TGFbeta in Cancer. Cell 2008, 134, 215–230. [Google Scholar] [CrossRef] [PubMed]
  93. Batlle, E.; Massagué, J. Transforming Growth Factor-β Signaling in Immunity and Cancer. Immunity 2019, 50, 924–940. [Google Scholar] [CrossRef]
  94. Ferrari, G.; Cook, B.D.; Terushkin, V.; Pintucci, G.; Mignatti, P. Transforming growth factor-beta 1 (TGF-β1) induces angiogenesis through vascular endothelial growth factor (VEGF)-mediated apoptosis. J. Cell. Physiol. 2009, 219, 449–458. [Google Scholar] [CrossRef]
  95. Chen, X.-H.; Liu, Z.-C.; Zhang, G.; Wei, W.; Wang, X.-X.; Wang, H.; Ke, H.-P.; Zhang, F.; Wang, H.-S.; Cai, S.-H.; et al. TGF-β and EGF induced HLA-I downregulation is associated with epithelial-mesenchymal transition (EMT) through upregulation of snail in prostate cancer cells. Mol. Immunol. 2015, 65, 34–42. [Google Scholar] [CrossRef] [PubMed]
  96. Zuber, P.; Kuppner, M.C.; De Tribolet, N. Transforming growth factor-β2 down-regulates HLA-DR antigen expression on human malignant glioma cells. Eur. J. Immunol. 1988, 18, 1623–1626. [Google Scholar] [CrossRef] [PubMed]
  97. Lebrin, F.; Deckers, M.; Bertolino, P.; Dijke, P.T. TGF-? receptor function in the endothelium. Cardiovasc. Res. 2005, 65, 599–608. [Google Scholar] [CrossRef] [PubMed]
  98. McKallip, R.; Li, R.; Ladisch, S. Tumor gangliosides inhibit the tumor-specific immune response. J. Immunol. 1999, 163, 3718–3726. [Google Scholar]
  99. Péguet-Navarro, J.; Sportouch, M.; Popa, I.; Berthier, O.; Schmitt, D.; Portoukalian, J. Gangliosides from Human Melanoma Tumors Impair Dendritic Cell Differentiation from Monocytes and Induce Their Apoptosis. J. Immunol. 2003, 170, 3488–3494. [Google Scholar] [CrossRef] [PubMed]
  100. Norment, A.M.; Salter, R.D.; Parham, P.; Engelhard, V.H.; Littman, D.R. Cell-cell adhesion mediated by CD8 and MHC class I molecules. Nature 1988, 336, 79–81. [Google Scholar] [CrossRef] [PubMed]
  101. Natarajan, K.; Li, H.; Mariuzza, R.A.; Margulies, D.H. MHC Class I Molecules, Structure and Function. Rev. Immunogenet. 1999, 1, 32–46. [Google Scholar] [PubMed]
  102. Movassaghi, M.; Chung, R.; Anderson, C.B.; Stein, M.; Saenger, Y.; Faiena, I. Overcoming Immune Resistance in Prostate Cancer: Challenges and Advances. Cancers 2021, 13, 4757. [Google Scholar] [CrossRef]
  103. Bander, N.H.; Yao, D.; Liu, H.; Chen, Y.-T.; Steiner, M.; Zuccaro, W.; Moy, P. MHC class I and II expression in prostate carcinoma and modulation by interferon-alpha and -gamma. Prostate 1997, 33, 233–239. [Google Scholar] [CrossRef]
  104. Ylitalo, E.B.; Thysell, E.; Jernberg, E.; Lundholm, M.; Crnalic, S.; Egevad, L.; Stattin, P.; Widmark, A.; Bergh, A.; Wikström, P. Subgroups of Castration-resistant Prostate Cancer Bone Metastases Defined Through an Inverse Relationship Between Androgen Receptor Activity and Immune Response. Eur. Urol. 2017, 71, 776–787. [Google Scholar] [CrossRef]
  105. Humeau, J.; Lévesque, S.; Kroemer, G.; Pol, J.G. Gold Standard Assessment of Immunogenic Cell Death in Oncological Mouse Models. Methods Mol. Biol. 2019, 1884, 297–315. [Google Scholar] [CrossRef]
  106. Bastianpillai, C.; Petrides, N.; Shah, T.; Guillaumier, S.; Ahmed, H.U.; Arya, M. Harnessing the immunomodulatory effect of thermal and non-thermal ablative therapies for cancer treatment. Tumor Biol. 2015, 36, 9137–9146. [Google Scholar] [CrossRef]
  107. Greenplate, A.R.; Johnson, D.B.; Ferrell, P.; Irish, J.M. Systems immune monitoring in cancer therapy. Eur. J. Cancer 2016, 61, 77–84. [Google Scholar] [CrossRef] [Green Version]
  108. Wolchok, J.D.; Neyns, B.; Linette, G.; Negrier, S.; Lutzky, J.; Thomas, L.; Waterfield, W.; Schadendorf, D.; Smylie, M.; Guthrie, T.; et al. Ipilimumab monotherapy in patients with pretreated advanced melanoma: A randomised, double-blind, multicentre, phase 2, dose-ranging study. Lancet Oncol. 2010, 11, 155–164. [Google Scholar] [CrossRef]
  109. Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved Survival with Ipilimumab in Patients with Metastatic Melanoma. N. Engl. J. Med. 2010, 363, 711. [Google Scholar] [CrossRef] [PubMed]
  110. Robert, C.; Schachter, J.; Long, G.V.; Arance, A.; Grob, J.J.; Mortier, L.; Daud, A.; Carlino, M.S.; McNeil, C.; Lotem, M.; et al. Pembrolizumab versus Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2015, 372, 2521–2532. [Google Scholar] [CrossRef]
  111. Huang, A.C.; Zappasodi, R. A decade of checkpoint blockade immunotherapy in melanoma: Understanding the molecular basis for immune sensitivity and resistance. Nat. Immunol. 2022, 23, 660–670. [Google Scholar] [CrossRef] [PubMed]
  112. Herbst, R.S.; Baas, P.; Kim, D.-W.; Felip, E.; Pérez-Gracia, J.L.; Han, J.-Y.; Molina, J.; Kim, J.-H.; Arvis, C.D.; Ahn, M.-J.; et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): A randomised controlled trial. Lancet 2015, 387, 1540–1550. [Google Scholar] [CrossRef]
  113. Brahmer, J.; Reckamp, K.L.; Baas, P.; Crinò, L.; Eberhardt, W.E.E.; Poddubskaya, E.; Antonia, S.; Pluzanski, A.; Vokes, E.E.; Holgado, E.; et al. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2015, 373, 123–135. [Google Scholar] [CrossRef] [PubMed]
  114. Carlino, M.S.; Larkin, J.; Long, G.V. Immune checkpoint inhibitors in melanoma. Lancet 2021, 398, 1002–1014. [Google Scholar] [CrossRef]
  115. Seidel, J.; 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]
  116. Hargadon, K.M.; Johnson, C.E.; Williams, C.J. Immune checkpoint blockade therapy for cancer: An overview of FDA-approved immune checkpoint inhibitors. Int. Immunopharmacol. 2018, 62, 29–39. [Google Scholar] [CrossRef]
  117. Darvin, P.; Toor, S.M.; Sasidharan Nair, V.; Elkord, E. Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp. Mol. Med. 2018, 50, 1–11. [Google Scholar] [CrossRef] [Green Version]
  118. Sharma, P.; Pachynski, R.K.; Narayan, V.; Fléchon, A.; Gravis, G.; Galsky, M.D.; Mahammedi, H.; Patnaik, A.; Subudhi, S.K.; Ciprotti, M.; et al. Nivolumab Plus Ipilimumab for Metastatic Castration-Resistant Prostate Cancer: Preliminary Analysis of Patients in the CheckMate 650 Trial. Cancer Cell 2020, 38, 489–499.e3. [Google Scholar] [CrossRef] [PubMed]
  119. Powles, T.; Yuen, K.C.; Gillessen, S.; Kadel, E.E.; Rathkopf, D.; Matsubara, N.; Drake, C.G.; Fizazi, K.; Piulats, J.M.; Wysocki, P.J.; et al. Atezolizumab with enzalutamide versus enzalutamide alone in metastatic castration-resistant prostate cancer: A randomized phase 3 trial. Nat. Med. 2022, 28, 144–153. [Google Scholar] [CrossRef]
  120. Agarwal, N.; Azad, A.; Carles, J.; Chowdhury, S.; McGregor, B.; Merseburger, A.S.; Oudard, S.; Saad, F.; Soares, A.; Benzaghou, F.; et al. A phase III, randomized, open-label study (CONTACT-02) of cabozantinib plus atezolizumab versus second novel hormone therapy in patients with metastatic castration-resistant prostate cancer. Futur. Oncol. 2022, 18, 1185–1198. [Google Scholar] [CrossRef]
  121. Mottet, N.; Cornford, P.; van den Bergh, R.C.N.; Briers, E.; De Santis, M.; Gillessen, S.; Grummet, J.; Henry, A.M.; van der Kwast, T.H.; Lam, T.B.; et al. EAU-EANM-ESTRO-ESUR-ISUP_SIOG Guidelines on Prostate Cancer 2022. Eur. Urol. 2022, 79, 243–262. [Google Scholar] [CrossRef] [PubMed]
  122. Le, D.T.; Uram, J.N.; Wang, H.; Bartlett, B.R.; Kemberling, H.; Eyring, A.D.; Skora, A.D.; Luber, B.S.; Azad, N.S.; Laheru, D.; et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N. Engl. J. Med. 2015, 372, 2509–2520. [Google Scholar] [CrossRef]
  123. Venkatachalam, S.; McFarland, T.; Agarwal, N.; Swami, U. Immune Checkpoint Inhibitors in Prostate Cancer. Cancers 2021, 13, 2187. [Google Scholar] [CrossRef]
  124. H, D.; Tf, H.; Ah, Z. The Current Role of Immunotherapy in mCRPC: A Systematic Review. Austin J. Clin. Case Rep. 2021, 8, 1222. [Google Scholar] [CrossRef]
  125. Karzai, F.; VanderWeele, D.; Madan, R.A.; Owens, H.; Cordes, L.M.; Hankin, A.; Couvillon, A.; Nichols, E.; Bilusic, M.; Beshiri, M.; et al. Activity of durvalumab plus olaparib in metastatic castration-resistant prostate cancer in men with and without DNA damage repair mutations. J. Immunother. Cancer 2018, 6, 141. [Google Scholar] [CrossRef]
  126. Antonarakis, E.S.; Velho, P.I.; Fu, W.; Wang, H.; Agarwal, N.; Santos, V.S.; Maughan, B.L.; Pili, R.; Adra, N.; Sternberg, C.N.; et al. CDK12-Altered Prostate Cancer: Clinical Features and Therapeutic Outcomes to Standard Systemic Therapies, Poly (ADP-Ribose) Polymerase Inhibitors, and PD-1 Inhibitors. JCO Precis. Oncol. 2020, 4, 370–381. [Google Scholar] [CrossRef]
  127. Graf, R.P.; Fisher, V.; Weberpals, J.; Gjoerup, O.; Tierno, M.B.; Huang, R.S.P.; Sayegh, N.; Lin, D.I.; Raskina, K.; Schrock, A.B.; et al. Comparative Effectiveness of Immune Checkpoint Inhibitors vs Chemotherapy by Tumor Mutational Burden in Metastatic Castration-Resistant Prostate Cancer. JAMA Netw. Open 2022, 5, e225394. [Google Scholar] [CrossRef] [PubMed]
  128. Vafaei, S.; Zekiy, A.O.; Khanamir, R.A.; Zaman, B.A.; Ghayourvahdat, A.; Azimizonuzi, H.; Zamani, M. Combination therapy with immune checkpoint inhibitors (ICIs); a new frontier. Cancer Cell Int. 2022, 22, 2. [Google Scholar] [CrossRef] [PubMed]
  129. Palmer, A.C.; Izar, B.; Hwangbo, H.; Sorger, P.K. Predictable Clinical Benefits without Evidence of Synergy in Trials of Combination Therapies with Immune-Checkpoint Inhibitors. Clin. Cancer Res. 2022, 28, 368–377. [Google Scholar] [CrossRef]
  130. Saxena, M.; van der Burg, S.H.; Melief, C.J.M.; Bhardwaj, N. Therapeutic cancer vaccines. Nat. Rev. Cancer 2021, 21, 360–378. [Google Scholar] [CrossRef]
  131. Abei, M.; Okumura, T.; Fukuda, K.; Hashimoto, T.; Araki, M.; Ishige, K.; Hyodo, I.; Kanemoto, A.; Numajiri, H.; Mizumoto, M.; et al. A phase I study on combined therapy with proton-beam radiotherapy and in situ tumor vaccination for locally advanced recurrent hepatocellular carcinoma. Radiat. Oncol. 2013, 8, 239. [Google Scholar] [CrossRef]
  132. Golden, E.B.; Chhabra, A.; Chachoua, A.; Adams, S.; Donach, M.; Fenton-Kerimian, M.; Friedman, K.; Ponzo, F.; Babb, J.S.; Goldberg, J.; et al. Local radiotherapy and granulocyte-macrophage colony-stimulating factor to generate abscopal responses in patients with metastatic solid tumours: A proof-of-principle trial. Lancet Oncol. 2015, 16, 795–803. [Google Scholar] [CrossRef]
  133. Kim, S.; A Kim, S.; Nam, G.-H.; Hong, Y.; Kim, G.B.; Choi, Y.; Lee, S.; Cho, Y.; Kwon, M.; Jeong, C.; et al. In situ immunogenic clearance induced by a combination of photodynamic therapy and rho-kinase inhibition sensitizes immune checkpoint blockade response to elicit systemic antitumor immunity against intraocular melanoma and its metastasis. J. Immunother. Cancer 2021, 9, e001481. [Google Scholar] [CrossRef] [PubMed]
  134. Lin, M.; Liang, S.; Wang, X.; Liang, Y.; Zhang, M.; Chen, J.; Niu, L.; Xu, K. Short-term clinical efficacy of percutaneous irreversible electroporation combined with allogeneic natural killer cell for treating metastatic pancreatic cancer. Immunol. Lett. 2017, 186, 20–27. [Google Scholar] [CrossRef]
  135. Schoenfeld, J.D.; Giobbie-Hurder, A.; Ranasinghe, S.; Kao, K.Z.; Lako, A.; Tsuji, J.; Liu, Y.; Brennick, R.C.; Gentzler, R.D.; Lee, C.; et al. Durvalumab plus tremelimumab alone or in combination with low-dose or hypofractionated radiotherapy in metastatic non-small-cell lung cancer refractory to previous PD(L)-1 therapy: An open-label, multicentre, randomised, phase 2 trial. Lancet Oncol. 2022, 23, 279–291. [Google Scholar] [CrossRef]
  136. DeMaria, P.J.; Bilusic, M.; Park, D.M.; Heery, C.R.; Donahue, R.N.; Madan, R.A.; Bagheri, M.H.; Strauss, J.; Shen, V.; Marté, J.L.; et al. Randomized, Double-Blind, Placebo-Controlled Phase II Study of Yeast-Brachyury Vaccine (GI-6301) in Combination with Standard-of-Care Radiotherapy in Locally Advanced, Unresectable Chordoma. Oncol. 2021, 26, e847–e858. [Google Scholar] [CrossRef] [PubMed]
  137. Sebastian, M.; Papachristofilou, A.; Weiss, C.; Früh, M.; Cathomas, R.; Hilbe, W.; Wehler, T.; Rippin, G.; Koch, S.D.; Scheel, B.; et al. Phase Ib study evaluating a self-adjuvanted mRNA cancer vaccine (RNActive®) combined with local radiation as consolidation and maintenance treatment for patients with stage IV non-small cell lung cancer. BMC Cancer 2014, 14, 748. [Google Scholar] [CrossRef] [Green Version]
  138. Gulley, J.L.; Arlen, P.M.; Bastian, A.; Morin, S.; Marte, J.; Beetham, P.; Tsang, K.-Y.; Yokokawa, J.; Hodge, J.W.; Ménard, C.; et al. Combining a Recombinant Cancer Vaccine with Standard Definitive Radiotherapy in Patients with Localized Prostate Cancer. Clin. Cancer Res. 2005, 11, 3353–3362. [Google Scholar] [CrossRef] [PubMed]
  139. Nam, J.; Son, S.; Park, K.S.; Moon, J.J. Photothermal Therapy Combined with Neoantigen Cancer Vaccination for Effective Immunotherapy against Large Established Tumors and Distant Metastasis. Adv. Ther. 2021, 4, 2100093. [Google Scholar] [CrossRef]
  140. Hwang, H.S.; Cherukula, K.; Bang, Y.J.; Vijayan, V.; Moon, M.J.; Thiruppathi, J.; Puth, S.; Jeong, Y.Y.; Park, I.-K.; Lee, S.E.; et al. Combination of Photodynamic Therapy and a Flagellin-Adjuvanted Cancer Vaccine Potentiated the Anti-PD-1-Mediated Melanoma Suppression. Cells 2020, 9, 2432. [Google Scholar] [CrossRef]
  141. Rodriguez-Ruiz, M.E.; Perez-Gracia, J.L.; Rodríguez, I.; Alfaro, C.; Oñate, C.; Pérez, G.; Gil-Bazo, I.; Benito, A.; Inogés, S.; López-Diaz de Cerio, A.; et al. Combined immunotherapy encompassing intratumoral poly-ICLC, dendritic-cell vaccination and radiotherapy in advanced cancer patients. Ann. Oncol. 2018, 29, 1312–1319. [Google Scholar] [CrossRef]
  142. Sadeghzadeh, M.; Bornehdeli, S.; Mohahammadrezakhani, H.; Abolghasemi, M.; Poursaei, E.; Asadi, M.; Zafari, V.; Aghebati-Maleki, L.; Shanehbandi, D. Dendritic cell therapy in cancer treatment; the state-of-the-art. Life Sci. 2020, 254, 117580. [Google Scholar] [CrossRef]
  143. Cheever, M.A.; Higano, C.S. PROVENGE (Sipuleucel-T) in Prostate Cancer: The First FDA-Approved Therapeutic Cancer Vaccine. Clin. Cancer Res. 2011, 17, 3520–3526. [Google Scholar] [CrossRef]
  144. Polo, S.H.; Muñoz, D.M.; Rodríguez, A.R.; Ruiz, J.S.; Rodríguez, D.R.; Couñago, F. Changing the History of Prostate Cancer with New Targeted Therapies. Biomedicines 2021, 9, 392. [Google Scholar] [CrossRef]
  145. George, D.J.; Nabhan, C.; DeVries, T.; Whitmore, J.B.; Gomella, L.G. Survival Outcomes of Sipuleucel-T Phase III Studies: Impact of Control-Arm Cross-Over to Salvage Immunotherapy. Cancer Immunol. Res. 2015, 3, 1063–1069. [Google Scholar] [CrossRef]
  146. Draube, A.; Klein-González, N.; Mattheus, S.; Brillant, C.; Hellmich, M.; Engert, A.; Von Bergwelt-Baildon, M. Dendritic Cell Based Tumor Vaccination in Prostate and Renal Cell Cancer: A Systematic Review and Meta-Analysis. PLoS ONE 2011, 6, e18801. [Google Scholar] [CrossRef]
  147. Fucikova, J.; Podrazil, M.; Jarolim, L.; Bilkova, P.; Hensler, M.; Becht, E.; Gasova, Z.; Klouckova, J.; Kayserova, J.; Horvath, R.; et al. Phase I/II trial of dendritic cell-based active cellular immunotherapy with DCVAC/PCa in patients with rising PSA after primary prostatectomy or salvage radiotherapy for the treatment of prostate cancer. Cancer Immunol. Immunother. 2017, 67, 89–100. [Google Scholar] [CrossRef] [PubMed]
  148. Vogelzang, N.J.; Beer, T.M.; Gerritsen, W.; Oudard, S.; Wiechno, P.; Kukielka-Budny, B.; Samal, V.; Hajek, J.; Feyerabend, S.; Khoo, V.; et al. Efficacy and Safety of Autologous Dendritic Cell–Based Immunotherapy, Docetaxel, and Prednisone vs Placebo in Patients With Metastatic Castration-Resistant Prostate Cancer. JAMA Oncol. 2022, 8, 546. [Google Scholar] [CrossRef]
  149. Xi, H.-B.; Wang, G.-X.; Fu, B.; Liu, W.-P.; Li, Y. Survivin and PSMA Loaded Dendritic Cell Vaccine for the Treatment of Prostate Cancer. Biol. Pharm. Bull. 2015, 38, 827–835. [Google Scholar] [CrossRef] [PubMed]
  150. Scheid, E.; Major, P.; Bergeron, A.; Finn, O.J.; Salter, R.D.; Eady, R.; Yassine-Diab, B.; Favre, D.; Peretz, Y.; Landry, C.; et al. Tn-MUC1 DC Vaccination of Rhesus Macaques and a Phase I/II Trial in Patients with Nonmetastatic Castrate-Resistant Prostate Cancer. Cancer Immunol. Res. 2016, 4, 881–892. [Google Scholar] [CrossRef] [PubMed]
  151. Castiello, L.; Sabatino, M.; Ren, J.; Terabe, M.; Khuu, H.; Wood, L.V.; Berzofsky, J.A.; Stroncek, D.F. Expression of CD14, IL10, and Tolerogenic Signature in Dendritic Cells Inversely Correlate with Clinical and Immunologic Response to TARP Vaccination in Prostate Cancer Patients. Clin. Cancer Res. 2017, 23, 3352–3364. [Google Scholar] [CrossRef] [PubMed]
  152. Wood, L.V.; Fojo, A.; Roberson, B.D.; Hughes, M.S.B.; Dahut, W.; Gulley, J.L.; Madan, R.A.; Arlen, P.M.; Sabatino, M.; Stroncek, D.F.; et al. TARP vaccination is associated with slowing in PSA velocity and decreasing tumor growth rates in patients with Stage D0 prostate cancer. OncoImmunology 2016, 5, e1197459. [Google Scholar] [CrossRef]
  153. Sanda, M.G.; Smith, D.C.; Charles, L.G.; Hwang, C.; Pienta, K.J.; Schlom, J.; Milenic, D.; Panicali, D.; Montie, J.E. Recombinant vaccinia-PSA (PROSTVAC) can induce a prostate-specific immune response in androgen-modulated human prostate cancer. Urology 1999, 53, 260–266. [Google Scholar] [CrossRef]
  154. Lasek, W.; Zapała, Ł. Therapeutic Metastatic Prostate Cancer Vaccines: Lessons Learnt from Urologic Oncology. Cent. Eur. J. Urol. 2021, 74, 300–307. [Google Scholar] [CrossRef]
  155. Sater, H.A.; Marté, J.L.; Donahue, R.N.; Walter-Rodriguez, B.; Heery, C.R.; Steinberg, S.M.; Cordes, L.M.; Chun, G.; Karzai, F.; Bilusic, M.; et al. Neoadjuvant PROSTVAC prior to radical prostatectomy enhances T-cell infiltration into the tumor immune microenvironment in men with prostate cancer. J. Immunother. Cancer 2019, 8, e000655. [Google Scholar] [CrossRef]
  156. Gulley, J.L.; Borre, M.; Vogelzang, N.J.; Ng, S.; Agarwal, N.; Parker, C.C.; Pook, D.; Rathenborg, P.; Flaig, T.W.; Carles, J.; et al. Phase III Trial of PROSTVAC in Asymptomatic or Minimally Symptomatic Metastatic Castration-Resistant Prostate Cancer. J. Clin. Oncol. 2019, 37, 1051–1061. [Google Scholar] [CrossRef]
  157. Parsons, J.K.; Pinto, P.A.; Pavlovich, C.P.; Uchio, E.; Kim, H.L.; Nguyen, M.N.; Gulley, J.L.; Jamieson, C.; Hsu, P.; Wojtowicz, M.; et al. A Randomized, Double-blind, Phase II Trial of PSA-TRICOM (PROSTVAC) in Patients with Localized Prostate Cancer: The Immunotherapy to Prevent Progression on Active Surveillance Study. Eur. Urol. Focus 2018, 4, 636–638. [Google Scholar] [CrossRef] [PubMed]
  158. Maiorano, B.; Schinzari, G.; Ciardiello, D.; Rodriquenz, M.; Cisternino, A.; Tortora, G.; Maiello, E. Cancer Vaccines for Genitourinary Tumors: Recent Progresses and Future Possibilities. Vaccines 2021, 9, 623. [Google Scholar] [CrossRef] [PubMed]
  159. Cappuccini, F.; Pollock, E.; Stribbling, S.; Hill, A.V.; Redchenko, I. 5T4 oncofoetal glycoprotein: An old target for a novel prostate cancer immunotherapy. Oncotarget 2017, 8, 47474–47489. [Google Scholar] [CrossRef]
  160. Fenoglio, D.; Traverso, P.; Parodi, A.; Tomasello, L.; Negrini, S.; Kalli, F.; Battaglia, F.; Ferrera, F.; Sciallero, M.S.; Murdaca, G.; et al. A multi-peptide, dual-adjuvant telomerase vaccine (GX301) is highly immunogenic in patients with prostate and renal cancer. Cancer Immunol. Immunother. 2013, 62, 1041–1052. [Google Scholar] [CrossRef] [PubMed]
  161. Filaci, G.; Fenoglio, D.; Nolè, F.; Zanardi, E.; Tomasello, L.; Aglietta, M.; Del Conte, G.; Carles, J.; Morales-Barrera, R.; Guglielmini, P.; et al. Telomerase-based GX301 cancer vaccine in patients with metastatic castration-resistant prostate cancer: A randomized phase II trial. Cancer Immunol. Immunother. 2021, 70, 3679–3692. [Google Scholar] [CrossRef]
  162. Obara, W.; Sato, F.; Takeda, K.; Kato, R.; Kato, Y.; Kanehira, M.; Takata, R.; Mimata, H.; Sugai, T.; Nakamura, Y.; et al. Phase I clinical trial of cell division associated 1 (CDCA1) peptide vaccination for castration resistant prostate cancer. Cancer Sci. 2017, 108, 1452–1457. [Google Scholar] [CrossRef]
  163. Obara, W.; Kanehira, M.; Katagiri, T.; Kato, R.; Kato, Y.; Takata, R. Present status and future perspective of peptide-based vaccine therapy for urological cancer. Cancer Sci. 2018, 109, 550–559. [Google Scholar] [CrossRef] [PubMed]
  164. Uemura, H.; Fujimoto, K.; Mine, T.; Uejima, S.; De Velasco, M.; Hirao, Y.; Komatsu, N.; Yamada, A.; Itoh, K. Immunological evaluation of personalized peptide vaccination monotherapy in patients with castration-resistant prostate cancer. Cancer Sci. 2010, 101, 601–608. [Google Scholar] [CrossRef]
  165. Noguchi, M.; Fujimoto, K.; Arai, G.; Uemura, H.; Hashine, K.; Matsumoto, H.; Fukasawa, S.; Kohjimoto, Y.; Nakatsu, H.; Takenaka, A.; et al. A randomized phase III trial of personalized peptide vaccination for castration-resistant prostate cancer progressing after docetaxel. Oncol. Rep. 2020, 45, 159–168. [Google Scholar] [CrossRef]
  166. Schuhmacher, J.; Heidu, S.; Balchen, T.; Richardson, J.R.; Schmeltz, C.; Sonne, J.; Schweiker, J.; Rammensee, H.-G.; Straten, P.T.; Røder, M.A.; et al. Vaccination against RhoC induces long-lasting immune responses in patients with prostate cancer: Results from a phase I/II clinical trial. J. Immunother. Cancer 2020, 8, e001157. [Google Scholar] [CrossRef] [PubMed]
  167. Santegoets, S.J.; Stam, A.G.M.; Lougheed, S.M.; Gall, H.; Jooss, K.; Sacks, N.; Hege, K.M.; Lowy, I.; Scheper, R.J.; Gerritsen, W.R.; et al. Myeloid derived suppressor and dendritic cell subsets are related to clinical outcome in prostate cancer patients treated with prostate GVAX and ipilimumab. J. Immunother. Cancer 2014, 2, 31. [Google Scholar] [CrossRef] [Green Version]
  168. Van den Eertwegh, A.J.M.; Versluis, J.; van den Berg, H.P.; Santegoets, S.J.A.M.; van Moorselaar, R.J.A.; van der Sluis, T.M.; Gall, H.E.; Harding, T.C.; Jooss, K.; Lowy, I.; et al. Combined immunotherapy with granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells and ipilimumab in patients with metastatic castration-resistant prostate cancer: A phase 1 dose-escalation trial. Lancet Oncol. 2012, 13, 509–517. [Google Scholar] [CrossRef]
  169. Karan, D.; Van Veldhuizen, P. Combination immunotherapy with prostate GVAX and ipilimumab: Safety and toxicity. Immunotherapy 2012, 4, 577–580. [Google Scholar] [CrossRef]
  170. Mottet, N.; Bastian, P.; Bellmunt, J.; van den Bergh, R.; Bolla, M.; van Casteren, N.; Cornford, P.; Joniau, S.; Matveev, V.; van der Kwast, T.; et al. EAU-EANM-ESTRO-ESUR-SIOG: Guidelines on Prostate Cancer. Eur. Assoc. Urol. 2020, 1–182. [Google Scholar]
  171. Beerlage, H.; Thüroff, S.; Debruyne, F.; Chaussy, C.; de la Rosette, J. Transrectal high-intensity focused ultrasound using the Ablatherm device in the treatment of localized prostate carcinoma. Urology 1999, 54, 273–277. [Google Scholar] [CrossRef]
  172. Gelet, A.; Chapelon, J.; Bouvier, R.; Rouvière, O.; Lasne, Y.; Lyonnet, D.; Dubernard, J. Transrectal High-Intensity Focused Ultrasound: Minimally Invasive Therapy of Localized Prostate Cancer. J. Endourol. 2000, 14, 519–528. [Google Scholar] [CrossRef] [PubMed]
  173. Ghai, S.; Finelli, A.; Corr, K.; Chan, R.; Jokhu, S.; Li, X.; McCluskey, S.; Konukhova, A.; Hlasny, E.; van der Kwast, T.H.; et al. MRI-guided Focused Ultrasound Ablation for Localized Intermediate-Risk Prostate Cancer: Early Results of a Phase II Trial. Radiology 2021, 298, 695–703. [Google Scholar] [CrossRef]
  174. Thüroff, S.; Chaussy, C.; Vallancien, G.; Wieland, W.; Kiel, H.J.; Le Duc, A.; Desgrandchamps, F.; De La Rosette, J.J.M.C.H.; Gelet, A. High-Intensity Focused Ultrasound and Localized Prostate Cancer: Efficacy Results from the European Multicentric Study. J. Endourol. 2003, 17, 673–677. [Google Scholar] [CrossRef]
  175. Uchida, T.; Baba, S.; Irie, A.; Soh, S.; Masumori, N.; Tsukamoto, T.; Nakatsu, H.; Fujimoto, H.; Kakizoe, T.; Ueda, T.; et al. Transrectal high-intensity focused ultrasound in the treatment of localized prostate cancer: A multicenter study. Acta Urol. Jpn. 2005, 51, 651–658. [Google Scholar]
  176. Koch, M.O.; Gardner, T.; Cheng, L.; Fedewa, R.J.; Seip, R.; Sangvhi, N.T. Phase I/II Trial of High Intensity Focused Ultrasound for the Treatment of Previously Untreated Localized Prostate Cancer. J. Urol. 2007, 178, 2366–2371. [Google Scholar] [CrossRef] [PubMed]
  177. Muto, S.; Yoshii, T.; Saito, K.; Kamiyama, Y.; Ide, H.; Horie, S. Focal Therapy with High-intensity-focused Ultrasound in the Treatment of Localized Prostate Cancer. Jpn. J. Clin. Oncol. 2008, 38, 192–199. [Google Scholar] [CrossRef] [PubMed]
  178. Crouzet, S.; Rebillard, X.; Chevallier, D.; Rischmann, P.; Pasticier, G.; Garcia, G.; Rouviere, O.; Chapelon, J.-Y.; Gelet, A. Multicentric Oncologic Outcomes of High-Intensity Focused Ultrasound for Localized Prostate Cancer in 803 Patients. Eur. Urol. 2010, 58, 559–566. [Google Scholar] [CrossRef]
  179. Crouzet, S.; Chapelon, J.Y.; Rouvière, O.; Mege-Lechevallier, F.; Colombel, M.; Tonoli-Catez, H.; Martin, X.; Gelet, A. Whole-gland Ablation of Localized Prostate Cancer with High-intensity Focused Ultrasound: Oncologic Outcomes and Morbidity in 1002 Patients. Eur. Urol. 2014, 65, 907–914. [Google Scholar] [CrossRef]
  180. Hamdy, F.C.; Elliott, D.; Le Conte, S.; Davies, L.C.; Burns, R.M.; Thomson, C.; Gray, R.; Wolstenholme, J.; Donovan, J.L.; Fitzpatrick, R.; et al. Partial ablation versus radical prostatectomy in intermediate-risk prostate cancer: The PART feasibility RCT. Health Technol. Assess. 2018, 22, 1–95. [Google Scholar] [CrossRef] [PubMed]
  181. Ganzer, R.; Hadaschik, B.; Pahernik, S.; Koch, D.; Baumunk, D.; Kuru, T.; Heidenreich, A.; Stolzenburg, J.-U.; Schostak, M.; Blana, A. Prospective Multicenter Phase II Study on Focal Therapy (Hemiablation) of the Prostate with High Intensity Focused Ultrasound. J. Urol. 2017, 199, 983–989. [Google Scholar] [CrossRef]
  182. Ramsay, C.R.; Adewuyi, E.T.; Gray, J.; Hislop, J.; Shirley, M.; Jayakody, S.; MacLennan, G.; Fraser, C.; MacLennan, S.; Brazzelli, M.; et al. Ablative therapy for people with localised prostate cancer: A systematic review and economic evaluation. Health Technol. Assess. 2015, 19, 1–8. [Google Scholar] [CrossRef]
  183. Schmid, F.; Schindele, D.; Mortezavi, A.; Spitznagel, T.; Sulser, T.; Schostak, M.; Eberli, D. Prospective multicentre study using high intensity focused ultrasound (HIFU) for the focal treatment of prostate cancer: Safety outcomes and complications. Urol. Oncol. Semin. Orig. Investig. 2019, 38, 225–230. [Google Scholar] [CrossRef] [PubMed]
  184. Joiner, J.B.; Pylayeva-Gupta, Y.; Dayton, P.A. Focused Ultrasound for Immunomodulation of the Tumor Microenvironment. J. Immunol. 2020, 205, 2327–2341. [Google Scholar] [CrossRef]
  185. Finley, D.S.; Pouliot, F.; Shuch, B.; Chin, A.; Pantuck, A.; Dekernion, J.B.; Belldegrun, A.S. Ultrasound-based combination therapy: Potential in urologic cancer. Expert Rev. Anticancer Ther. 2011, 11, 107–113. [Google Scholar] [CrossRef]
  186. Ter Haar, G.; Coussios, C. High intensity focused ultrasound: Physical principles and devices. Int. J. Hyperth. 2007, 23, 89–104. [Google Scholar] [CrossRef]
  187. Hu, Z.; Yang, X.Y.; Liu, Y.; Sankin, G.N.; Pua, E.C.; A Morse, M.; Lyerly, H.K.; Clay, T.M.; Zhong, P. Investigation of HIFU-induced anti-tumor immunity in a murine tumor model. J. Transl. Med. 2007, 5, 34. [Google Scholar] [CrossRef]
  188. Ran, L.-F.; Xie, X.-P.; Xia, J.-Z.; Xie, F.-L.; Fan, Y.-M.; Wu, F. Specific antitumour immunity of HIFU-activated cytotoxic T lymphocytes after adoptive transfusion in tumour-bearing mice. Int. J. Hyperth. 2015, 32, 204–210. [Google Scholar] [CrossRef] [PubMed]
  189. Rosberger, D.F.; Coleman, D.J.; Silverman, R.; Woods, S.; Rondeau, M.; Cunningham-Rundles, S. Immunomodulation in Choroidal Melanoma: Reversal of Inverted CD4/CD8 Ratios Following Treatment with Ultrasonic Hyperthermia. Biotechnol. Ther. 1994, 5, 59–68. [Google Scholar]
  190. Zhang, Y.; Deng, J.; Feng, J.; Wu, F. Enhancement of antitumor vaccine in ablated hepatocellular carcinoma by high-intensity focused ultrasound. World J. Gastroenterol. 2010, 16, 3584–3591. [Google Scholar] [CrossRef] [PubMed]
  191. 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]
  192. 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]
  193. Yang, R.; Reilly, C.R.; Rescorla, F.J.; Sanghvi, N.T.; Fry, F.J.; Franklin, T.D.; Grosfeld, J.L. Effects of high-intensity focused ultrasound in the treatment of experimental neuroblastoma. J. Pediatr. Surg. 1992, 27, 246–251. [Google Scholar] [CrossRef]
  194. Eranki, A.; Srinivasan, P.; Ries, M.; Kim, A.; Lazarski, C.A.; Rossi, C.T.; Khokhlova, T.D.; Wilson, E.; Knoblach, S.M.; Sharma, K.V.; et al. High-Intensity Focused Ultrasound (HIFU) Triggers Immune Sensitization of Refractory Murine Neuroblastoma to Checkpoint Inhibitor Therapy. Clin. Cancer Res. 2020, 26, 1152–1161. [Google Scholar] [CrossRef] [PubMed]
  195. Wang, X.; Sun, J. High-intensity focused ultrasound in patients with late-stage pancreatic carcinoma. Chin. Med. J. 2002, 115, 1332–1335. [Google Scholar]
  196. Mouratidis, P.X.E.; Costa, M.; Rivens, I.; Repasky, E.E.; ter Haar, G. Pulsed focused ultrasound can improve the anti-cancer effects of immune checkpoint inhibitors in murine pancreatic cancer. J. R. Soc. Interface 2021, 18, 20210266. [Google Scholar] [CrossRef] [PubMed]
  197. Wu, F.; Wang, Z.-B.; Cao, Y.-D.; Zhou, Q.; Zhang, Y.; Xu, Z.-L.; Zhu, X.-Q. Expression of Tumor Antigens and Heat-Shock Protein 70 in Breast Cancer Cells After High-Intensity Focused Ultrasound Ablation. Ann. Surg. Oncol. 2006, 14, 1237–1242. [Google Scholar] [CrossRef]
  198. Ji, C.; Si, J.; Xu, Y.; Zhang, W.; Yang, Y.; He, X.; Xu, H.; Mou, X.; Ren, H.; Guo, H. Mitochondria-targeted and ultrasound-responsive nanoparticles for oxygen and nitric oxide codelivery to reverse immunosuppression and enhance sonodynamic therapy for immune activation. Theranostics 2021, 11, 8587–8604. [Google Scholar] [CrossRef] [PubMed]
  199. Zhang, D.; Lin, Z.; Zheng, Y.; Song, J.; Li, J.; Zeng, Y.; Liu, X. Ultrasound-Driven Biomimetic Nanosystem Suppresses Tumor Growth and Metastasis through Sonodynamic Therapy, CO Therapy, and Indoleamine 2,3-Dioxygenase Inhibition. ACS Nano 2020, 14, 8985–8999. [Google Scholar] [CrossRef]
  200. Wen, J.; Duan, Y.; Zou, Y.; Nie, Z.; Feng, H.; Lugnani, F.; Baust, J.G. Cryoablation induces necrosis and apoptosis in lung adenocarcinoma in mice. Technol. Cancer Res. Treat. 2007, 6, 635–640. [Google Scholar] [CrossRef] [PubMed]
  201. Robilotto, A.T.; Baust, J.M.; Van Buskirk, R.G.; Gage, A.A.; Baust, J.G. Temperature-dependent activation of differential apoptotic pathways during cryoablation in a human prostate cancer model. Prostate Cancer Prostatic Dis. 2012, 16, 41–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Kimura, M.; Rabbani, Z.; Mouraviev, V.; Tsivian, M.; Vujaskovic, Z.; Satoh, T.; Baba, S.; Baust, J.M.; Polascik, T.J. Morphology of hypoxia following cryoablation in a prostate cancer murine model: Its relationship to necrosis, apoptosis and, microvessel density. Cryobiology 2010, 61, 148–154. [Google Scholar] [CrossRef] [PubMed]
  203. Yakkala, C.; Denys, A.; Kandalaft, L.; Duran, R. Cryoablation and immunotherapy of cancer. Curr. Opin. Biotechnol. 2020, 65, 60–64. [Google Scholar] [CrossRef] [PubMed]
  204. Aarts, B.M.; Klompenhouwer, E.G.; Rice, S.L.; Imani, F.; Baetens, T.; Bex, A.; Horenblas, S.; Kok, M.; Haanen, J.B.A.G.; Beets-Tan, R.G.H.; et al. Cryoablation and immunotherapy: An overview of evidence on its synergy. Insights Imaging 2019, 10, 53. [Google Scholar] [CrossRef] [PubMed]
  205. Zhu, J.; Zhang, Y.; Zhang, A.; He, K.; Liu, P.; Xu, L.X. Cryo-thermal therapy elicits potent anti-tumor immunity by inducing extracellular Hsp70-dependent MDSC differentiation. Sci. Rep. 2016, 6, 27136. [Google Scholar] [CrossRef]
  206. Erinjeri, J.P.; Clark, T.W. Cryoablation: Mechanism of Action and Devices. J. Vasc. Interv. Radiol. 2010, 21, S187–S191. [Google Scholar] [CrossRef]
  207. Ablin, R.J.; Soanes, A.W.; Gonder, M.J. Immunologic studies of the prostate. A review. Int. Surg. 1969, 52, 8–21. [Google Scholar] [PubMed]
  208. Abdo, J.; Cornell, D.L.; Mittal, S.K.; Agrawal, D.K. Immunotherapy Plus Cryotherapy: Potential Augmented Abscopal Effect for Advanced Cancers. Front. Oncol. 2018, 8, 85. [Google Scholar] [CrossRef]
  209. Stamell, E.F.; Wolchok, J.D.; Gnjatic, S.; Lee, N.Y.; Brownell, I. The Abscopal Effect Associated With a Systemic Anti-melanoma Immune Response. Int. J. Radiat. Oncol. 2012, 85, 293–295. [Google Scholar] [CrossRef] [PubMed]
  210. Gaitanis, G.; Bassukas, I.D. Immunocryosurgery for non-superficial basal cell carcinomas ≤ 20 mm in maximal diameter: Five-year follow-up. J. Geriatr. Oncol. 2018, 10, 475–478. [Google Scholar] [CrossRef] [PubMed]
  211. Lin, M.; Xu, K.; Liang, S.; Wang, X.; Liang, Y.; Zhang, M.; Chen, J.; Niu, L. Prospective study of percutaneous cryoablation combined with allogenic NK cell immunotherapy for advanced renal cell cancer. Immunol. Lett. 2017, 184, 98–104. [Google Scholar] [CrossRef]
  212. Lin, M.; Liang, S.; Wang, X.; Liang, Y.; Zhang, M.; Chen, J.; Niu, L.; Xu, K. Cryoablation combined with allogenic natural killer cell immunotherapy improves the curative effect in patients with advanced hepatocellular cancer. Oncotarget 2017, 8, 81967–81977. [Google Scholar] [CrossRef]
  213. Lin, M.; Liang, S.-Z.; Wang, X.-H.; Liang, Y.-Q.; Zhang, M.-J.; Niu, L.-Z.; Chen, J.-B.; Li, H.-B.; Xu, K.-C. Clinical efficacy of percutaneous cryoablation combined with allogenic NK cell immunotherapy for advanced non-small cell lung cancer. Immunol. Res. 2017, 65, 880–887. [Google Scholar] [CrossRef] [PubMed]
  214. Si, T.; Guo, Z.; Hao, X. Combined Cryoablation and GM-CSF Treatment for Metastatic Hormone Refractory Prostate Cancer. J. Immunother. 2009, 32, 86–91. [Google Scholar] [CrossRef]
  215. Barqawi, A.B.; Pessoa, R.R.; Crawford, E.D.; Al-Musawi, M.; MacDermott, T.; O’Donell, C.; Kendl, R.M. Boosting immune response with GM-CSF optimizes primary cryotherapy outcomes in the treatment of prostate cancer: A prospective randomized clinical trial. Prostate Cancer Prostatic Dis. 2021, 24, 750–757. [Google Scholar] [CrossRef] [PubMed]
  216. Ross, A.E.; Hurley, P.J.; Tran, P.T.; Rowe, S.P.; Benzon, B.; Neal, T.O.; Chapman, C.; Harb, R.; Milman, Y.; Trock, B.J.; et al. A pilot trial of pembrolizumab plus prostatic cryotherapy for men with newly diagnosed oligometastatic hormone-sensitive prostate cancer. Prostate Cancer Prostatic Dis. 2019, 23, 184–193. [Google Scholar] [CrossRef]
  217. Wang, G.; Zhao, D.; Spring, D.J.; Depinho, R.A. Genetics and biology of prostate cancer. Genes Dev. 2018, 32, 1105–1140. [Google Scholar] [CrossRef]
  218. Zhou, C. New trends in photobiology: Mechanisms of tumor necrosis induced by photodynamic therapy. J. Photochem. Photobiol. B: Biol. 1989, 3, 299–318. [Google Scholar] [CrossRef]
  219. Wang, M.; Rao, J.; Wang, M.; Li, X.; Liu, K.; Naylor, M.F.; Nordquist, R.E.; Chen, W.R.; Zhou, F. Cancer photo-immunotherapy: From bench to bedside. Theranostics 2021, 11, 2218–2231. [Google Scholar] [CrossRef]
  220. Algorri, J.F.; Ochoa, M.; Roldán-Varona, P.; Rodríguez-Cobo, L.; López-Higuera, J.M. Photodynamic Therapy: A Compendium of Latest Reviews. Cancers 2021, 13, 4447. [Google Scholar] [CrossRef]
  221. Rastinehad, A.R.; Anastos, H.; Wajswol, E.; Winoker, J.S.; Sfakianos, J.P.; Doppalapudi, S.K.; Carrick, M.R.; Knauer, C.J.; Taouli, B.; Lewis, S.C.; et al. Gold nanoshell-localized photothermal ablation of prostate tumors in a clinical pilot device study. Proc. Natl. Acad. Sci. USA 2019, 116, 18590–18596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Azzouzi, A.-R.; Vincendeau, S.; Barret, E.; Cicco, A.; Kleinclauss, F.; van der Poel, H.G.; Stief, C.G.; Rassweiler, J.; Salomon, G.; Solsona, E.; et al. Padeliporfin vascular-targeted photodynamic therapy versus active surveillance in men with low-risk prostate cancer (CLIN1001 PCM301): An open-label, phase 3, randomised controlled trial. Lancet Oncol. 2016, 18, 181–191. [Google Scholar] [CrossRef]
  223. Osuchowski, M.; Bartusik-Aebisher, D.; Osuchowski, F.; Aebisher, D. Photodynamic therapy for prostate cancer—A narrative review. Photodiagnosis Photodyn. Ther. 2020, 33, 102158. [Google Scholar] [CrossRef]
  224. Sandler, H. FDA Committee Votes against Approval of Tookad for Localized Prostate Cancer. Available online: https://www.healio.com/news/hematology-oncology/20200226/fda-committee-votes-against-approval-of-tookad-for-localized-prostate-cancer (accessed on 12 August 2022).
  225. Castano, A.P.; Mroz, P.; Hamblin, M.R. Photodynamic therapy and anti-tumour immunity. Nat. Cancer 2006, 6, 535–545. [Google Scholar] [CrossRef] [PubMed]
  226. Yang, G.; Ni, J.; Li, Y.; Zha, M.; Tu, Y.; Li, K. Acceptor Engineering for Optimized ROS Generation Facilitates Reprogramming Macrophages to M1 Phenotype in Photodynamic Immunotherapy. Angew. Chem. Int. Ed. 2020, 60, 5386–5393. [Google Scholar] [CrossRef] [PubMed]
  227. Hu, J.; Song, J.; Tang, Z.; Wei, S.; Chen, L.; Zhou, R. Hypericin-mediated photodynamic therapy inhibits growth of colorectal cancer cells via inducing S phase cell cycle arrest and apoptosis. Eur. J. Pharmacol. 2021, 900, 174071. [Google Scholar] [CrossRef] [PubMed]
  228. Zhang, D.; Xie, Q.; Liu, Y.; Li, Z.; Li, H.; Li, S.; Li, Z.; Cui, J.; Su, M.; Jiang, X.; et al. Photosensitizer IR700DX-6T- and IR700DX-mbc94-mediated photodynamic therapy markedly elicits anticancer immune responses during treatment of pancreatic cancer. Pharmacol. Res. 2021, 172, 105811. [Google Scholar] [CrossRef]
  229. Theodoraki, M.; Lorenz, K.; Lotfi, R.; Fürst, D.; Tsamadou, C.; Jaekle, S.; Mytilineos, J.; Brunner, C.; Theodorakis, J.; Hoffmann, T.; et al. Influence of photodynamic therapy on peripheral immune cell populations and cytokine concentrations in head and neck cancer. Photodiagnosis Photodyn. Ther. 2017, 19, 194–201. [Google Scholar] [CrossRef]
  230. Hwang, H.S.; Shin, H.; Han, J.; Na, K. Combination of photodynamic therapy (PDT) and anti-tumor immunity in cancer therapy. J. Pharm. Investig. 2018, 48, 143–151. [Google Scholar] [CrossRef] [PubMed]
  231. Zou, J.; Li, L.; Yang, Z.; Chen, X. Phototherapy meets immunotherapy: A win–win strategy to fight against cancer. Nanophotonics 2021, 10, 3229–3245. [Google Scholar] [CrossRef]
  232. Kabingu, E.; Vaughan, L.; Owczarczak, B.; Ramsey, K.D.; O Gollnick, S. CD8+ T cell-mediated control of distant tumours following local photodynamic therapy is independent of CD4+ T cells and dependent on natural killer cells. Br. J. Cancer 2007, 96, 1839–1848. [Google Scholar] [CrossRef] [Green Version]
  233. Mroz, P.; Hashmi, J.T.; Huang, Y.; Lange, N.; Hamblin, M.R. Stimulation of anti-tumor immunity by photodynamic therapy. Expert Rev. Clin. Immunol. 2011, 7, 75–91. [Google Scholar] [CrossRef] [PubMed]
  234. Li, Y.; Li, X.; Doughty, A.; West, C.; Wang, L.; Zhou, F.; Nordquist, R.E.; Chen, W.R. Phototherapy using immunologically modified carbon nanotubes to potentiate checkpoint blockade for metastatic breast cancer. Nanomed. Nanotechnol. Biol. Med. 2019, 18, 44–53. [Google Scholar] [CrossRef] [PubMed]
  235. Huang, Z.; Wei, G.; Zeng, Z.; Huang, Y.; Huang, L.; Shen, Y.; Sun, X.; Xu, C.; Zhao, C. Enhanced cancer therapy through synergetic photodynamic/immune checkpoint blockade mediated by a liposomal conjugate comprised of porphyrin and IDO inhibitor. Theranostics 2019, 9, 5542–5557. [Google Scholar] [CrossRef] [PubMed]
  236. Nagaya, T.; Nakamura, Y.; Okuyama, S.; Ogata, F.; Maruoka, Y.; Choyke, P.L.; Kobayashi, H. Near-Infrared Photoimmunotherapy Targeting Prostate Cancer with Prostate-Specific Membrane Antigen (PSMA) Antibody. Mol. Cancer Res. 2017, 15, 1153–1162. [Google Scholar] [CrossRef] [PubMed]
  237. Watanabe, R.; Hanaoka, H.; Kobayashi, H.; Sato, K.; Nagaya, T.; Harada, T.; Mitsunaga, M.; Kim, I.; Paik, C.H.; Wu, A.M.; et al. Photoimmunotherapy targeting prostate-specific membrane antigen: Are antibody fragments as effective as antibodies? J. Nucl. Med. 2015, 56, 140–144. [Google Scholar] [CrossRef]
  238. Miller, L.; Leor, J.; Rubinsky, B. Cancer Cells Ablation with Irreversible Electroporation. Technol. Cancer Res. Treat. 2005, 4, 699–705. [Google Scholar] [CrossRef] [PubMed]
  239. Scheltema, M.J.V.; Bos, W.V.D.; De Bruin, D.M.; Wijkstra, H.; Laguna, M.P.; De Reijke, T.M.; De La Rosette, J.J.M.C.H. Focal vs extended ablation in localized prostate cancer with irreversible electroporation; a multi-center randomized controlled trial. BMC Cancer 2016, 16, 299. [Google Scholar] [CrossRef] [PubMed]
  240. Bos, W.V.D.; Jurhill, R.R.; De Bruin, D.M.; Heijink, D.S.; Postema, A.W.; Wagstaff, P.G.K.; Muller, B.G.; Varkarakis, I.M.; Skolarikos, A.; Zondervan, P.J.; et al. Histopathological Outcomes after Irreversible Electroporation for Prostate Cancer: Results of an Ablate and Resect Study. J. Urol. 2016, 196, 552–559. [Google Scholar] [CrossRef] [PubMed]
  241. Bos, W.V.D.; De Bruin, D.M.; Van Randen, A.; Engelbrecht, M.R.W.; Postema, A.; Muller, B.G.; Varkarakis, I.M.; Skolarikos, A.; Heijink, D.S.; Jurhill, R.R.; et al. MRI and contrast-enhanced ultrasound imaging for evaluation of focal irreversible electroporation treatment: Results from a phase I-II study in patients undergoing IRE followed by radical prostatectomy. Eur. Radiol. 2015, 26, 2252–2260. [Google Scholar] [CrossRef] [PubMed]
  242. Valerio, M.; Dickinson, L.; Ali, A.; Ramachadran, N.; Donaldson, I.; McCartan, N.; Freeman, A.; Ahmed, H.U.; Emberton, M. Nanoknife Electroporation Ablation Trial: A Prospective Development Study Investigating Focal Irreversible Electroporation for Localized Prostate Cancer. J. Urol. 2017, 197, 647–654. [Google Scholar] [CrossRef] [PubMed]
  243. Collettini, F.; Enders, J.; Stephan, C.; Fischer, T.; Baur, A.D.; Penzkofer, T.; Busch, J.; Hamm, B.; Gebauer, B. Image-guided Irreversible Electroporation of Localized Prostate Cancer: Functional and Oncologic Outcomes. Radiology 2019, 292, 250–257. [Google Scholar] [CrossRef]
  244. Bos, W.V.D.; Scheltema, M.J.; Siriwardana, A.R.; Kalsbeek, A.M.; Thompson, J.E.; Ting, F.; Böhm, M.; Haynes, A.-M.; Shnier, R.; Delprado, W.; et al. Focal irreversible electroporation as primary treatment for localized prostate cancer. Br. J. Urol. 2017, 121, 716–724. [Google Scholar] [CrossRef]
  245. Dong, S.; Wang, H.; Zhao, Y.; Sun, Y.; Yao, C. First Human Trial of High-Frequency Irreversible Electroporation Therapy for Prostate Cancer. Technol. Cancer Res. Treat. 2018, 17, 1–9. [Google Scholar] [CrossRef] [PubMed]
  246. Wang, H.; Xue, W.; Yan, W.; Yin, L.; Dong, B.; He, B.; Yu, Y.; Shi, W.; Zhou, Z.; Lin, H.; et al. Extended Focal Ablation of Localized Prostate Cancer with High-Frequency Irreversible Electroporation: An Objective Performance Criteria Trial. JAMA Surg. 2022, 157, 693–700. [Google Scholar] [CrossRef] [PubMed]
  247. Ting, F.; Tran, M.; Böhm, M.; Siriwardana, A.; Van Leeuwen, P.J.; Haynes, A.-M.; Delprado, W.; Shnier, R.; Stricker, P. Focal irreversible electroporation for prostate cancer: Functional outcomes and short-term oncological control. Prostate Cancer Prostatic Dis. 2015, 19, 46–52. [Google Scholar] [CrossRef]
  248. Kiełbik, A.; Szlasa, W.; Saczko, J.; Kulbacka, J. Electroporation-Based Treatments in Urology. Cancers 2020, 12, 2208. [Google Scholar] [CrossRef] [PubMed]
  249. Yang, J.; Eresen, A.; Shangguan, J.; Ma, Q.; Yaghmai, V.; Zhang, Z. Irreversible electroporation ablation overcomes tumor-associated immunosuppression to improve the efficacy of DC vaccination in a mice model of pancreatic cancer. OncoImmunology 2021, 10, 1875638. [Google Scholar] [CrossRef]
  250. Zhao, J.; Wen, X.; Tian, L.; Li, T.; Xu, C.; Wen, X.; Melancon, M.P.; Gupta, S.; Shen, B.; Peng, W.; et al. Irreversible electroporation reverses resistance to immune checkpoint blockade in pancreatic cancer. Nat. Commun. 2019, 10, 899. [Google Scholar] [CrossRef] [PubMed]
  251. He, C.; Sun, S.; Zhang, Y.; Li, S. Irreversible Electroporation Plus Anti-PD-1 Antibody versus Irreversible Electroporation Alone for Patients with Locally Advanced Pancreatic Cancer. J. Inflamm. Res. 2021, 14, 4795–4807. [Google Scholar] [CrossRef] [PubMed]
  252. Burbach, B.J.; O’Flanagan, S.D.; Shao, Q.; Young, K.M.; Slaughter, J.R.; Rollins, M.R.; Street, T.J.L.; Granger, V.E.; Beura, L.K.; Azarin, S.M.; et al. Irreversible electroporation augments checkpoint immunotherapy in prostate cancer and promotes tumor antigen-specific tissue-resident memory CD8+ T cells. Nat. Commun. 2021, 12, 3862. [Google Scholar] [CrossRef]
  253. Eriksson, D.; Stigbrand, T. Radiation-induced cell death mechanisms. Tumor Biol. 2010, 31, 363–372. [Google Scholar] [CrossRef] [PubMed]
  254. Finkelstein, S.E.; Salenius, S.; Mantz, C.A.; Shore, N.D.; Fernandez, E.B.; Shulman, J.; Myslicki, F.A.; Agassi, A.M.; Rotterman, Y.; DeVries, T.; et al. Combining Immunotherapy and Radiation for Prostate Cancer. Clin. Genitourin. Cancer 2015, 13, 1–9. [Google Scholar] [CrossRef] [PubMed]
  255. Nesslinger, N.J.; Sahota, R.A.; Stone, B.; Johnson, K.; Chima, N.; King, C.; Rasmussen, D.; Bishop, D.; Rennie, P.S.; Gleave, M.; et al. Standard Treatments Induce Antigen-Specific Immune Responses in Prostate Cancer. Clin. Cancer Res. 2007, 13, 1493–1502. [Google Scholar] [CrossRef] [PubMed]
  256. Lin, L.; Kane, N.; Kobayashi, N.; Kono, E.A.; Yamashiro, J.M.; Nickols, N.G.; Reiter, R.E. High-dose per Fraction Radiotherapy Induces Both Antitumor Immunity and Immunosuppressive Responses in Prostate Tumors. Clin. Cancer Res. 2021, 27, 1505–1515. [Google Scholar] [CrossRef] [PubMed]
  257. De Martino, M.; Daviaud, C.; Vanpouille-Box, C. Radiotherapy: An immune response modifier for immuno-oncology. Semin. Immunol. 2021, 52, 101474. [Google Scholar] [CrossRef]
  258. Donlon, N.; Power, R.; Hayes, C.; Reynolds, J.; Lysaght, J. Radiotherapy, immunotherapy, and the tumour microenvironment: Turning an immunosuppressive milieu into a therapeutic opportunity. Cancer Lett. 2021, 502, 84–96. [Google Scholar] [CrossRef]
  259. Solanki, A.A.; Bossi, A.; Efstathiou, J.A.; Lock, D.; Mondini, M.; Ramapriyan, R.; Welsh, J.; Kang, J. Combining Immunotherapy with Radiotherapy for the Treatment of Genitourinary Malignancies. Eur. Urol. Oncol. 2018, 2, 79–87. [Google Scholar] [CrossRef]
  260. Demaria, S.; Bhardwaj, N.; McBride, W.H.; Formenti, S.C. Combining radiotherapy and immunotherapy: A revived partnership. Int. J. Radiat. Oncol. 2005, 63, 655–666. [Google Scholar] [CrossRef] [PubMed]
  261. Eckert, F.; Schaedle, P.; Zips, D.; Schmid-Horch, B.; Rammensee, H.-G.; Gani, C.; Gouttefangeas, C. Impact of curative radiotherapy on the immune status of patients with localized prostate cancer. OncoImmunology 2018, 7, e1496881. [Google Scholar] [CrossRef] [PubMed]
  262. Nickols, N.G.; Ganapathy, E.; Nguyen, C.; Kane, N.; Lin, L.; Diaz-Perez, S.; Nazarian, R.; Mathis, C.; Felix, C.; Basehart, V.; et al. The intraprostatic immune environment after stereotactic body radiotherapy is dominated by myeloid cells. Prostate Cancer Prostatic Dis. 2020, 24, 135–139. [Google Scholar] [CrossRef] [PubMed]
  263. Dudzinski, S.O.; Cameron, B.D.; Wang, J.; Rathmell, J.C.; Giorgio, T.D.; Kirschner, A.N. Combination immunotherapy and radiotherapy causes an abscopal treatment response in a mouse model of castration resistant prostate cancer. J. Immunother. Cancer 2019, 7, 218. [Google Scholar] [CrossRef]
  264. Wada, S.; Harris, T.J.; Tryggestad, E.; Yoshimura, K.; Zeng, J.; Yen, H.-R.; Getnet, D.; Grosso, J.F.; Bruno, T.C.; De Marzo, A.M.; et al. Combined Treatment Effects of Radiation and Immunotherapy: Studies in an Autochthonous Prostate Cancer Model. Int. J. Radiat. Oncol. 2013, 87, 769–776. [Google Scholar] [CrossRef] [PubMed]
  265. Philippou, Y.; Sjoberg, H.T.; Murphy, E.; Alyacoubi, S.; Jones, K.I.; Gordon-Weeks, A.N.; Phyu, S.; Parkes, E.E.; McKenna, W.G.; Lamb, A.D.; et al. Impacts of combining anti-PD-L1 immunotherapy and radiotherapy on the tumour immune microenvironment in a murine prostate cancer model. Br. J. Cancer 2020, 123, 1089–1100. [Google Scholar] [CrossRef] [PubMed]
  266. Slovin, S.F.; Higano, C.S.; Hamid, O.; Tejwani, S.; Harzstark, A.; Alumkal, J.J.; Scher, H.I.; Chin, K.; Gagnier, P.; McHenry, M.B.; et al. Ipilimumab alone or in combination with radiotherapy in metastatic castration-resistant prostate cancer: Results from an open-label, multicenter phase I/II study. Ann. Oncol. 2013, 24, 1813–1821. [Google Scholar] [CrossRef] [PubMed]
  267. Kwon, E.D.; Drake, C.G.; Scher, H.I.; Fizazi, K.; Bossi, A.; Van den Eertwegh, A.J.M.; Krainer, M.; Houede, N.; Santos, R.; Mahammedi, H.; et al. Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): A multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol. 2014, 15, 700–712. [Google Scholar] [CrossRef]
  268. Twardowski, P.; Wong, J.Y.; Pal, S.K.; Maughan, B.L.; Frankel, P.H.; Franklin, K.; Junqueira, M.; Prajapati, M.R.; Nachaegari, G.; Harwood, D.; et al. Randomized phase II trial of sipuleucel-T immunotherapy preceded by sensitizing radiation therapy and sipuleucel-T alone in patients with metastatic castrate resistant prostate cancer. Cancer Treat. Res. Commun. 2018, 19, 100116. [Google Scholar] [CrossRef] [PubMed]
  269. Yuan, Z.; Fernandez, D.; Dhillon, J.; Abraham-Miranda, J.; Awasthi, S.; Kim, Y.; Zhang, J.; Jain, R.; Serna, A.; Pow-Sang, J.M.; et al. Proof-of-principle Phase I results of combining nivolumab with brachytherapy and external beam radiation therapy for Grade Group 5 prostate cancer: Safety, feasibility, and exploratory analysis. Prostate Cancer Prostatic Dis. 2020, 24, 140–149. [Google Scholar] [CrossRef] [PubMed]
  270. Marshall, C.H.; Fu, W.; Wang, H.; Park, J.C.; DeWeese, T.L.; Tran, P.T.; Song, D.Y.; King, S.; Afful, M.; Hurrelbrink, J.; et al. Randomized Phase II Trial of Sipuleucel-T with or without Radium-223 in Men with Bone-metastatic Castration-resistant Prostate Cancer. Clin. Cancer Res. 2021, 27, 1623–1630. [Google Scholar] [CrossRef]
  271. Han, H.J.; Li, Y.R.; Roach, I.M.; Aggarwal, R. Dramatic response to combination pembrolizumab and radiation in metastatic castration resistant prostate cancer. Ther. Adv. Med. Oncol. 2020, 12, 1758835920936084. [Google Scholar] [CrossRef]
  272. Noack, M.; Beringer, A.; Miossec, P. Additive or Synergistic Interactions Between IL-17A or IL-17F and TNF or IL-1β Depend on the Cell Type. Front. Immunol. 2019, 10, 1726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Prado, D.S.; Cattley, R.T.; Shipman, C.W.; Happe, C.; Lee, M.; Boggess, W.C.; MacDonald, M.L.; Hawse, W.F. Synergistic and additive interactions between receptor signaling networks drive the regulatory T cell versus T helper 17 cell fate choice. J. Biol. Chem. 2021, 297, 101330. [Google Scholar] [CrossRef] [PubMed]
  274. Sheng, X.; Nenseth, H.Z.; Qu, S.; Kuzu, O.F.; Frahnow, T.; Simon, L.; Greene, S.; Zeng, Q.; Fazli, L.; Rennie, P.S.; et al. IRE1α-XBP1s pathway promotes prostate cancer by activating c-MYC signaling. Nat. Commun. 2019, 10, 323. [Google Scholar] [CrossRef] [PubMed]
  275. Amin, M.; Gao, F.; Terrero, G.; Picus, J.; Wang-Gillam, A.; Suresh, R.; Ma, C.; Tan, B.; Baggstrom, M.; Naughton, M.J.; et al. Phase I Study of Docetaxel and Temsirolimus in Refractory Solid Tumors. Am. J. Clin. Oncol. 2021, 44, 443–448. [Google Scholar] [CrossRef] [PubMed]
  276. Wei, M.-M.; Zhao, S.-J.; Dong, X.-M.; Wang, Y.-J.; Fang, C.; Wu, P.; Song, G.-Q.; Gao, J.-N.; Huang, Z.-H.; Xie, T.; et al. A combination index and glycoproteomics-based approach revealed synergistic anticancer effects of curcuminoids of turmeric against prostate cancer PC3 cells. J. Ethnopharmacol. 2020, 267, 113467. [Google Scholar] [CrossRef] [PubMed]
  277. Palmer, A.C.; Sorger, P.K. Combination Cancer Therapy Can Confer Benefit via Patient-to-Patient Variability without Drug Additivity or Synergy. Cell 2017, 171, 1678–1691.e13. [Google Scholar] [CrossRef] [PubMed]
  278. Geary, N. Understanding synergy. Am. J. Physiol. Metab. 2013, 304, E237–E253. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic overview of cancer immunity cycle.
Figure 1. Schematic overview of cancer immunity cycle.
Cells 11 02793 g001
Table 1. Ongoing trials assessing combinations of radiotherapy and immunotherapy.
Table 1. Ongoing trials assessing combinations of radiotherapy and immunotherapy.
NCT NumberPhasenSettingImmunotherapeuticsRadiotherapy
NCT03835533I45mCRPCNKTR-214, Nivolumab, CDX-301, Poly-ICLC, INO-5151SBRT
NCT03795207II96mPCDurvalumabSBRT
NCT03543189I/II44PCNivolumabBrachytherapy, EBRT
NCT03217747I/II173mCRPCAnti-OX40, Avelumab, UtomilumabRT *
NCT03007732II42PCPembrolizumab, SD-101SBRT
NCT01818986II20mCRPCSipuleucel-TSBRT
NCT01436968III711PCAglatimagene BesadenovecEBRT
NCT: The National Clinical Trial; n: number of patients enrolled; PC: prostate cancer; mPC: metastatic prostate cancer; mCRPC: metastatic castration-resistant prostate cancer; RT: radiotherapy; SBRT: stereotactic body radiation therapy; EBRT: external beam radiation therapy. * The specific variant of radiation therapy was not specified.
Table 2. Immunomodulatory impact of local treatment strategies on the TME.
Table 2. Immunomodulatory impact of local treatment strategies on the TME.
Local TherapyImmunomodulatory EffectsReferences
HIFU
Promotion of DCs infiltration and activation.
Increase in CD3+ and CD4+ levels cells, and CD4+/CD8+ ratio.
Enhancement of CTLs cytotoxicity.
[187,188,189,190,191,192,193,194,195,196,197,198,199]
Cryotherapy
Activation of T cells.
DC maturation.
The abscopal effect.
[204,207,208,209,210,211,212,213,214,215,216]
PDT
Promotion of neutrophils recruitment.
DC maturation.
Activation of macrophages.
Regulation of CTL and NK cell migration, increase in CD8+ T cell infiltration.
Secretion of IL-1, IL-6, IL-8, IL-10, IL-12, thromboxane, and prostaglandins.
[133,225,230,231,232,233,234,235,236,237]
IRE
Decrease in Tregs and MDSC levels.
[248,249,250,251,252]
RT
Enhancement of damage-associated molecule release.
Activation of DCs and other APCs.
Release of various cytokines (e.g., CXC10, CXCL16, IL-1, TNF-α, and interferons).
Upregulation of MHC I, NKG2D ligand and Fas/CD95.
The abscopal effect.
[253,258,259,260,261,262,263,264,265,266,267,268,269,270,271]
HIFU: high-intensity focused ultrasound; PDT: photodynamic therapy; IRE: irreversible electroporation; RT: radiotherapy; DC: dendritic cell; CTL: cytotoxic T lymphocyte; NK: natural killer; IL: interleukin; Treg: regulatory T cell; MDSC: myeloid-derived suppressor cell; APC: antigen-presenting cell; TNF: tumor necrosis factor; MHC: major histocompatibility complex.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Karwacki, J.; Kiełbik, A.; Szlasa, W.; Sauer, N.; Kowalczyk, K.; Krajewski, W.; Saczko, J.; Kulbacka, J.; Szydełko, T.; Małkiewicz, B. Boosting the Immune Response—Combining Local and Immune Therapy for Prostate Cancer Treatment. Cells 2022, 11, 2793. https://doi.org/10.3390/cells11182793

AMA Style

Karwacki J, Kiełbik A, Szlasa W, Sauer N, Kowalczyk K, Krajewski W, Saczko J, Kulbacka J, Szydełko T, Małkiewicz B. Boosting the Immune Response—Combining Local and Immune Therapy for Prostate Cancer Treatment. Cells. 2022; 11(18):2793. https://doi.org/10.3390/cells11182793

Chicago/Turabian Style

Karwacki, Jakub, Aleksander Kiełbik, Wojciech Szlasa, Natalia Sauer, Kamil Kowalczyk, Wojciech Krajewski, Jolanta Saczko, Julita Kulbacka, Tomasz Szydełko, and Bartosz Małkiewicz. 2022. "Boosting the Immune Response—Combining Local and Immune Therapy for Prostate Cancer Treatment" Cells 11, no. 18: 2793. https://doi.org/10.3390/cells11182793

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