Next Article in Journal
Vitamin D and Mitosis Evaluation in Endometriosis: A Step toward Discovering the Connection?
Next Article in Special Issue
Evaluation of the Effectiveness of an Innovative Polycomponent Formulation on Adult and Aged Human Dermal Fibroblasts
Previous Article in Journal
Advancements and Insights in Exosome-Based Therapies for Wound Healing: A Comprehensive Systematic Review (2018–June 2023)
Previous Article in Special Issue
Serum Urate Levels and Ultrasound Characteristics of Carotid Atherosclerosis across Obesity Phenotypes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Holistic Approach to Hard-to-Treat Cancers: The Future of Immunotherapy for Glioblastoma, Triple Negative Breast Cancer, and Advanced Prostate Cancer

by
Carles Puig-Saenz
1,2,
Joshua R. D. Pearson
1,2,
Jubini E. Thomas
1,2 and
Stéphanie E. B. McArdle
1,2,*
1
The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, College Drive, Clifton, Nottingham NG11 8NS, UK
2
Centre for Systems Health and Integrated Metabolic Research, School of Science and Technology, Nottingham Trent University, College Drive, Clifton, Nottingham NG11 8NS, UK
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(8), 2100; https://doi.org/10.3390/biomedicines11082100
Submission received: 27 June 2023 / Revised: 21 July 2023 / Accepted: 24 July 2023 / Published: 25 July 2023

Abstract

:
Immunotherapy represents an attractive avenue for cancer therapy due to its tumour specificity and relatively low frequency of adverse effects compared to other treatment modalities. Despite many advances being made in the field of cancer immunotherapy, very few immunotherapeutic treatments have been approved for difficult-to-treat solid tumours such as triple negative breast cancer (TNBC), glioblastoma multiforme (GBM), and advanced prostate cancer (PCa). The anatomical location of some of these cancers may also make them more difficult to treat. Many trials focus solely on immunotherapy and have failed to consider or manipulate, prior to the immunotherapeutic intervention, important factors such as the microbiota, which itself is directly linked to lifestyle factors, diet, stress, social support, exercise, sleep, and oral hygiene. This review summarises the most recent treatments for hard-to-treat cancers whilst factoring in the less conventional interventions which could tilt the balance of treatment in favour of success for these malignancies.

Graphical Abstract

1. Introduction

Immunotherapy, such as the use of checkpoint inhibitors, oncolytic virus, bispecific antibodies, and adoptive cell transfer, has revolutionised the treatment of many cancers. Today, many of these approaches have been FDA approved for the treatment of several cancers [1]. Talimogene laherparepvec (T-VEC) is an oncolytic virus, FDA approved in 2015, for the treatment of advanced melanoma (stage IIIB-IV) [2] and Mosunetuzumab-axgb is a bispecific antibody approved for the treatment of adults with relapsed or refractory follicular lymphoma; in addition, three other bispecific antibodies have recently been approved for the treatment of acute lymphoblastic leukaemia (Blincyto), for non-small cell lung cancer (Rybrevant), and for uveal melanoma (Kimmtrak). In terms of cellular therapy, FDA-approved treatment using chimeric antigen receptor T (CAR-T) has been limited to B cell malignancies expressing CD19 and, while these harbour high potential for solid tumours, they also have significant toxicity, including severe cytokine release syndrome (CRS) and substantial neurotoxicity. Additionally, PROVENGE is the only FDA-approved immunotherapy for the treatment of advanced prostate cancer (PCa) without DNA mismatch repair deficiency (dMMR); however, while the therapy is well tolerated, it remains expensive and limited in its efficacy. A vast number of additional approaches (neoantigens with adjuvants, dendritic cell (DC) vaccines, peptide/mRNA vaccines…) are being investigated, including any combinations of the aforementioned, FDA-approved treatments.
The immune system is known to be strongly affected by both intrinsic (age, sex, and genetics) and extrinsic (environmental and lifestyle) factors. While one cannot change an individual’s intrinsic factors, external factors, especially those linked to lifestyle, can be manipulated. Yet very few studies have attempted to combine “conventional approaches” with any of these. In this review, we summarise the difficulties faced when treating glioblastoma (GBM), triple negative breast cancer (TNBC), and PCa, and how environmental and lifestyle factors represent emerging parameters that strongly influence the progression of these diseases. We also discuss how some of these parameters have the potential to be added prior to and along with conventional therapy to increase overall survival and/or quality of life for the patients.

2. Glioblastoma (GBM)

GBM is the most frequently occurring primary brain tumour with an incidence rate of ~3 in 100,000. It affects both children and adults, although it is primarily a disease associated with increased age, with the average age of diagnosis being ~65 years. GBM carries a poor prognosis and is nearly always fatal, with only around 3–5% of patients surviving for a period of five years or more [3,4]. Current therapy involves surgical resection (where possible) followed by concomitant radiotherapy and temozolomide chemotherapy. Despite aggressive multimodal therapy, nearly all tumours recur close to the site of resection. Complete surgical resection is almost impossible due to the highly infiltrative nature of GBM [5]. In addition, not all tumours are responsive to temozolomide chemotherapy, and some patients’ tumours may express the enzyme O6-methylguanine-DNA-methyltransferase (MGMT). MGMT repairs the DNA damage induced by temozolomide, making patients whose tumours express MGMT resistant [6]. Very few new therapies have been approved for GBM in recent years and, due to the poor prognosis associated with GBM, new therapeutic interventions are desperately required. Immunotherapy represents an attractive therapy option due to its tumour specificity and the ability of activated immune cells to access the brain and target intracranial tumours. Numerous immunotherapeutic approaches are being trialled in the GBM setting (Table 1); however, currently there are no approved immunotherapies for GBM. While numerous successes have been seen in other cancers, as yet no immunotherapy has been approved for use in GBM, with several therapies failing to show efficacy during phase 3 testing (e.g., Rindopepimut) [7]. This could be due to a highly immunosuppressive tissue microenvironment, the inability of large molecules to cross the blood–brain barrier and penetrate tumours, and the low mutation rates compared to other tumours. T-cell dysfunction is also frequently seen in the GBM setting with cells often expressing exhaustion markers and having an altered metabolism [8,9,10]. Standard therapies used to manage GBM are also known to dampen the immune response, with one such example being corticosteroids used to treat GBM-associated oedema.
Research has begun to highlight the importance of several lifestyle factors when utilising the immune system to fight GBM. The gut microbiome has been identified as an important component of the immune system and a predictor of response to immunotherapy in the cancer setting. Indeed, the gut microbiome has been shown to be a predictor of response to anti-PD-1 immune checkpoint blockade in the murine GBM setting, with the presence of Bacteroides cellulosilyticus being linked with response to anti-PD-1 immune checkpoint therapy [65].
The alteration of patient diets has been examined as a therapeutic intervention in the cancer setting [66]. Several studies have looked at utilising a ketogenic diet to treat GBM with the aim of reducing carbohydrate intake and therefore starving the tumour cells of glucose. Furthermore, it seems as if a ketogenic diet can enhance immunity in a murine GBM model. Mice harbouring intracranial GL261-Luc2 tumours were given a ketogenic diet and it was found that these mice had a significant reduction in immune inhibitory receptors such as PD-1 and CTLA-4 among their tumour-infiltrating lymphocytes [67]. This information points to a potential combinatorial role for the ketogenic diet with active immunotherapy.
Exercise also appears to have a potential role in the therapy of GBM. A case study reported that exercise improved the quality of life for a patient undergoing radiation treatment and she displayed signs of increased physical fitness such as muscle strength, balance, and aerobic capacity [68]. These impacts on the quality of life can also improve the psychological status of the patient, which may also have a knock-on effect on the anti-tumour immune response. Furthermore, irisin, a myokine associated with exercise, was shown to have anti-GBM properties in vitro, leading to cell cycle arrest in a panel of three cell lines. In vivo injection of irisin into a U-87 MG tumour led to a reduction in tumour size compared to untreated tumours and, even more impressively, mice that exercised on a running wheel also had reduced tumour growth compared to controls [69].
Several clinical trials investigate the effects of exercise, diet, and the microbiome on GBM (Table 2). Many of these holistic approaches have been examined as potential therapies on their own; however, several of these therapies could be used with active immunotherapy to boost the anti-tumour immune response and improve patient outcomes.

3. Triple Negative Breast Cancer (TNBC)

TNBC is a hard-to-treat type of breast cancer characterised by the lack of oestrogen receptor alpha (ERa), progesterone (PR), and HER2 receptors. Around 15% of all breast cancers fall under the category of triple negative [85], are more prevalent in black women, premenopausal women, women under 40 years of age, and women carrying BRCA1 mutations [86], and are extremely rare in men [87]. According to the Surveillance, Epidemiology, and End Result Program (SEER) database, TNBC bears an overall 5-year survival rate of 77% which drops down to 12% when the disease is at the metastatic stage [88]. In non-metastatic TNBC, between 30 and 40% of cases will result in relapse often leading to metastasis [89,90].
Compared to other types of breast cancer, the treatment options for TNBC are limited due to the lack of targetable receptors. ERa, PR, and HER2 are well known to play critical roles in the tumorigenesis of breast cancers, acting as therapeutic targets for a large proportion of patients [91,92]. Conventional treatments for TNBC include breast-conserving surgery and mastectomy, usually followed by radiotherapy and/or chemotherapy. Common systemic agents include anthracyclines, platinum-based drugs, and taxanes [93]. Interestingly, despite the overall aggressiveness of TNBC, a significant proportion of patients achieve a pathologic complete response following neoadjuvant chemotherapy [94]; however, incomplete responses are associated with high risk of recurrence [95].
Due to the high heterogeneity of TNBC tumours, finding a common targeted therapy for all TNBC becomes challenging. Although none have yet been approved, some targeted therapies currently under investigation include poly (ADP-ribose) polymerase (PARP) inhibitors, which induce cell death in cells with BRCA mutations; androgen receptor antagonists, which stunt the growth of TNBC subtypes expressing androgen receptor; antiangiogenic agents such as vascular endothelial growth factor receptor (VEGFR) inhibitors, which block the recruitment of new blood vessels towards the tumour; and epigenetic regulators such as DNA methyltransferase and histone deacetylase inhibitors, which have shown the capacity to induce the expression of oestrogen receptors, sensitising tumours to hormone therapy [96]. In addition to these therapies, many vaccines are being trialled for the treatment of TNBC (Table 3).
Disruptions in the balanced diversity of the microbiota, referred to as microbiome dysbiosis, is known to contribute to several health disorders [113,114]. Advances in meta-omics research technologies are facilitating our understanding of how this phenomenon can lead to other less understood disorders, including cancer [115]. Additionally, attention is increasingly being paid to the modulating effects of the microbiome on treatments such as cancer immunotherapies [116]. Although not yet elucidated, increasing evidence suggests that dysbiosis may contribute to the pathogenesis of breast cancer in various ways [117]. Overall, the consensus is that cancers are associated with a reduced diversity in gut microbiota [118], with studies suggesting that breast cancer is not an exception [119,120,121].
Oestrogen metabolism is a potential mechanism by which the gut microbiome can influence breast cancer pathogenesis. Whether endogenous or exogenous, oestrogen is a known risk factor for breast cancer, particularly in postmenopausal women [122]. The gut microbiome seems to play a role in oestrogen-driven breast cancers by enzymatically deconjugating oestrogen, therefore forcing it back into circulation and increasing systemic levels [123]. In TNBC, however, the negative impact of microbiota-mediated oestrogen abundance does not seem to occur, likely due to the lack of ERa expression. TNBCs have alternative oestrogen signalling pathways which render them responsive to circulating hormones [124]. Interestingly, studies suggest that receptors such as ERb, G protein-coupled oestrogen receptor 1, and oestrogen-related receptors have anti-cancer effects and that, when these are expressed in TNBCs, prognosis seems to be better [125].
Moreover, the microbiome does not extend exclusively to the gastrointestinal tract; in fact, different parts of the human body have been found to host different populations of microbes, and these can vary among and within individuals due to factors such as diet, lifestyle, usage of antibiotics, and even social interactions [126]. It is therefore not surprising that there exists a breast tissue-specific microbiome [127]. Studies such as that of Tzeng et al. have found that the most abundant phylum of bacteria in both healthy and cancerous breast tissue is Proteobacteria, while TNBC tissue was composed of bacteria from the genera Azomonas, Alkanindiges, Caulobacter, Proteus, Brevibacillus, Kocurla, and Parasediminibacterium [128]. However, previous studies have identified different genera in TNBC tissues [128,129,130,131], highlighting the need for further research and refinement of methodologies to study the microbiome.
Physical exercise has long been known to offer a wide range of health benefits [132]. Among these benefits, it is said to act as an “immune system adjuvant” which improves the recirculation and activity of certain immune components [133]. Preclinical evidence suggests that lifestyle can have a positive effect on the immune system when it comes to fighting cancer. Hojman et al. demonstrated a reduction in tumour growth in mice with access to voluntary wheel running, highlighting an increase in tumour immune recognition by macrophages, NK and T cells, but a decreased recognition in mice fed high-fat diets [134]. In a model of TNBC, Wennerberg et al. found a reduction in tumour-induced myeloid-derived suppressor cell (MDSC) recruitment, as well as an increase in NK and CD8+ T cell activation in the exercise treatment group, including an improvement of response to PD-1 inhibition [135]. Obesity has a tumorigenic effect in TNBC. For example, it alters the immune response by reprogramming mammary adipose tissue macrophages to a pro-inflammatory metabolically activated phenotype [136]. It also contributes to metabolic dysregulation, with evidence suggesting that exercise can reduce tumour growth by means of metabolic—mitochondrial and macronutrient—regulation [137].
Clinical studies suggest that an improvement in overall and disease-free survival is observed following moderate physical exercise upon diagnosis [138,139], with obesity playing a negative role in the outcome of all subtypes of breast cancer [140]. Exercise may exert these effects in different ways. For example, decreasing kynurenine pathway metabolites [141]; this pathway is known to be dysregulated in TNBC, contributing to the inhibition of anti-tumour responses [142]. It may also have a positive impact on inflammatory cytokines [143] known to play a role in TNBC [144]. As the evidence mounts, it seems sensible to use the benefits of maintaining a healthy lifestyle in order to help prevent—and possibly be considered before and during immunotherapeutic interventions against—cancer. The World Cancer Research Fund and the American Institute for Cancer Research have published recommendations for cancer prevention, which include guidelines for physical activity as well as for healthy diets and weight [145]. Although further research is warranted, studies suggest that lifestyle changes have the potential to improve treatment response and risk of relapse [146], and clinical trials are being conducted to assess the effect of exercise, diet, and the microbiome in TNBC patients (Table 4).

4. Advanced Prostate Cancer (PCa)

PCa is one of the most diagnosed fatal malignancies among men worldwide [151]. Compared to other common cancers, the aetiology of PCa remains unknown. Advanced age, positive family history, prostate inflammation, obesity, lack of exercise, ethnicity, and persistent elevated levels of testosterone are some of the risk factors known for PCa [152].
Localised prostate cancer is primarily managed through active surveillance, radical prostatectomy, external radiotherapy, and brachytherapy [152]. However, there is evidence of biochemical recurrence of malignancy observed within ten years of initial treatment in approximately 30–50% of patients who received radiotherapy or 20–40% of patients who underwent prostatectomy [151]. The advanced stage of prostate cancer is typically treated with androgen deprivation therapy (ADT), which is effective in controlling cancer growth. However, most patients eventually progress to metastatic castration-resistant prostate cancer (mCRPC). The loss of testosterone resulting from ADT is often associated with intense side effects, including mood swings, erectile dysfunction, and loss of bone density. Other approved therapies for PCa, such as radium-223 and taxane chemotherapy, have shown limited improvement in overall survival for patients, as the cancer continues to progress [153].
Sipuleucel-T is the only FDA-approved cellular immunotherapy for PCa. This approach has shown to increase PCa patients’ overall survival by 4.1 months. This vaccine is prepared by collecting the patient’s peripheral blood mononuclear cells (PBMCs) through leukapheresis. The collected cells are then incubated ex vivo with PA2024, which is a recombinant fusion protein combining prostatic acid phosphatase (PAP) and granulocyte macrophage colony-stimulating factor (GM-CSF). Finally, the engineered product is reinfused back into the patient. The PAP antigen is specific to prostate tissue and is expressed in most prostate adenocarcinomas. However, the high cost of this treatment limits its widespread availability [154].
Immune checkpoint inhibitor treatments such as anti-PD1 have shown significant clinical benefit for PCa patients whose tumours harbour DNA mismatch repair deficiency (dMMR). However, these only account for 3–5% of all castration-resistant prostate cancer, and only have modest activity in unselected men with metastatic prostate cancer. It is highly likely that the limited clinical response of immunotherapy in PCa is due to the immunosuppressive tumour microenvironment (TME) associated with it. This environment is characterised by the presence of immunosuppressive cells such as tumour-associated macrophages, MDSCs, and regulatory T cells. Additionally, adenosine produced via PAP and transforming growth factor-β (TGF-β) act as potent immunosuppressive molecules [153]. Interestingly, among genitourinary malignancies, PCa exhibits a distinct TME profile. PCa-associated tumour intrinsic factors such as decreased MHC class I expression, low tumour-associated antigen expression, loss of tumour suppressor protein PTEN, dysfunctional signalling of type I interferons, and mutations in the DNA damage repair genes BRCA1 and BRCA2 contribute towards the evolution of immunologically cold PCa TME [155]. Furthermore, PCa biopsy samples have shown the presence of tumour-infiltrating lymphocytes (TILs) that are biased toward T-regulatory (Treg) and T-helper 17 (Th17) phenotypes, which suppress autoreactive T cells and anti-tumour immune responses [154]. Several immunotherapy trials for PCa are underway or have already been completed (Table 5).
Certain host factors such as composition of the gut microbiota may also facilitate PCa progression and impact response to chemotherapy and immunotherapy [154,162]. Occurrence of certain microorganisms such as Cutibacterium in human prostate can cause immunosuppression and prostatitis by stimulating the infiltration of CD4+FoxP3+ cells (Treg) and Th17 cells [162]. The composition of the gut microbiota also plays a significant role in the response elicited by ADT with its immunostimulatory or immunosuppressive and direct ADT subversion. Depletion of immunostimulatory gut microbiota by orally administered broad spectrum antibiotics in mouse models have been shown to diminish the efficacy of ADT [163].
The decreased levels of androgen due to ADT in PCa patients have been suggested to contribute to a reduction in α and β-diversity in gut microbiota, leading to the development of dysbiosis [163,164,165]. A study involving sequential faecal and blood samples collected from 23 PCa patients showed a significant difference in the abundance and composition of microbiota, including increased levels of Proteobacteria, Pseudomonas, and Gammaproteobacteria, after ADT compared to before ADT [164]. Certain intestinal bacteria have the ability to degrade ADT-relevant drugs, thereby reducing the effectiveness of the therapy. Specific gut microbiota can act as androgen-producing bacteria by converting androgen precursors into active androgen. The abundance of such microbiota has been observed in castrated mice as well as in patients with castration-resistant prostate cancer (CRPC). Interestingly, a significant reduction in circulating testosterone levels has been observed in castrated mice when their gut microbiota is depleted [163].
The abundance of gut microbiota that can interfere with the clinical responses to ADT ultimately leads to the development of CRPC [165]. In a study conducted by Liu and Jiang, the faecal microbiota of 21 patients who received ADT was profiled, revealing compositional differences in gut microbiota between hormone-sensitive prostate cancer and CRPC [165]. CRPC was found to have a significant increase in the abundance of fourteen phylotypes of microbial flora, including Phascolarctobacterium and Ruminococcus. Additionally, bacterial gene pathways involved in terpenoid/polyketide metabolism and ether lipid metabolism were notably activated in CRPC. Similarly, another study by Che et al. on faecal microbiota demonstrated significant differences in the abundance of bacteria between prostate cancer patients and healthy individuals, with metabolic pathways associated with folic acid and arginine being affected [166]. Folic acid is crucial for nucleotide synthesis and DNA methylation, and its deficiency can lead to DNA instability and increased mutation rates. Moreover, folic acid-producing microflora were found to be less abundant in PCa patients compared to non-cancer patients, suggesting that natural sources of folic acid may offer protection against prostate cancer [166].
Another contributing factor to intestinal dysbiosis is lifestyle, including factors such as diet and obesity, which are often associated with an increase in circulating levels of pro-inflammatory bacterial lipopolysaccharide (LPS), leading to the development of prostate cancer [162,163]. In mouse models, the accumulation of LPS has been shown to activate local inflammation and promote prostate tumour growth [162]. A diet high in saturated fat can also promote the progression of prostate cancer by increasing circulating levels of androgens and causing DNA damage in cells through elevated oxidative stress [167]. Additionally, a Western-style high-fat diet, which often leads to obesity, can induce chronic inflammation, and contribute to the development of prostate cancer by upregulating inflammatory cytokines such as IL-6 [163]. Clinical trials are looking at the effects of exercise, diet, and the microbiome on PCa (Table 6).
In addition to lifestyle factors, antibiotic exposure can also contribute to gut dysbiosis. Research conducted by Zhong et al. demonstrated that antibiotic exposure leads to an enrichment of gut Proteobacteria, increased gut permeability, and elevated levels of intra-tumoral lipopolysaccharide (LPS), which promote the development of prostate cancer through the NF-κB-IL6-STAT3 axis in mice [178].
Another interesting factor to consider is the signalling of β-adrenergic receptors (β-AR), which plays a vital role in the progression and metastasis of many cancers, including PCa [179]. Higher expression of β2-AR has been observed in carcinoma compared to normal prostate tissues, as observed in tissue microarray studies using immunohistochemistry [180]. In line with this finding, Zang et al. have uncovered a significant role for β2-AR signalling in regulating the activity of the Shh pathway in PCa tumorigenesis using xenograft models [180]. Moreover, the administration of propranolol, a nonselective β-AR blocker, has demonstrated anti-cancer effects in cancer cell lines and animal models [179].
A patient cohort study utilising the Taiwan National Health Insurance Research Database, covering the period from January 2000 to December 2011, examined the usage of propranolol in various cancers, including PCa. The study concluded that propranolol can reduce the risk of cancers, with the most substantial protective effect observed when propranolol usage exceeded 1000 days [179]. To understand the efficacy of β-blockers in PCa patient mortality, Grytli et al. conducted a study involving 3561 PCa patients, out of which 1115 patients used β-blockers before and after diagnosis [181]. The study found a reduction in cancer-specific mortality among high-risk or metastatic PCa patients who used β-blockers.
Considering the potential therapeutic options suggested by targeting gut microbiota dysbiosis [164,165] and β2-adrenergic modulation [180], a combination of these approaches with other available therapeutic strategies could potentially benefit the management of PCa.

5. Chronic Stress and Cancer

Acute stress is a beneficial neuroendocrine response to external or internal stressor events which in turn activates our fight-or-flight response [182]. On the other hand, chronic stress is known to have detrimental effects on various aspects of physiology, with increasing research suggesting that it could lead to cancer progression [183]. The stress response begins when the amygdala perceives danger stimuli and relays this information to the hypothalamus which, in turn, promotes the release of catecholamines from the adrenal glands—a neuroendocrine component known as the sympathetic–adreno–medullar axis (Figure 1). In the TME, adrenaline and noradrenaline can have an immunosuppressive effect during chronic stress, for example, by increasing MDSC frequency [184]. In addition, stress-induced dopamine has recently been involved in tumour progression via activation of hypoxia-inducible factor-1α [185]. The hypothalamus also produces corticotropin-releasing factor, which acts upon the pituitary gland to promote the release of adrenocorticotropic hormone; this hormone then travels to the adrenal cortex to stimulate the synthesis and secretion of corticosteroids in what is called the hypothalamus–pituitary–adrenal axis. Corticosteroids are widely known for their immunosuppressive effects, affecting key effectors of anti-tumour immunity such as dendritic cells and T cells [186] (Figure 1).
It is not uncommon for patients living with cancer and other life-threatening diseases to feel stressed, anxious, and/or depressed. Given that these negative mental health states can worsen cancer prognosis, it seems appropriate to pay more attention to patient wellbeing; for example, offering patients resilience-enhancing interventions during key stages of their disease could accelerate their mental health recovery [187]. Moreover, research points to various links between diet and depression, for example, the consumption of ultra-processed foods [188] as well as diets rich in proline; with regards to the latter, Mayneris-Perxachs et al. recently found that a healthy gut microbiome is associated with lower plasma proline levels and lower depression scores, highlighting the importance of maintaining a balanced diet [189]. In addition, Valles-Colomer et al. found a positive association between the presence of Dialister and Coprococcus species and quality of life, with these species being sparse in depressed individuals [190]. Given the interplay between mental health, the immune system, diet, the microbiome, and cancer (Figure 1), it seems sensible, therefore, to approach the mental wellbeing of patients from a holistic point of view.

6. Conclusions and Future Directions for Immunotherapy

Our immune system needs to be able to respond appropriately to external and internal environmental changes caused by factors such as physical or psychological stress, nutrient availability, the microbiota, temperature, pathogens, and malignancies. To achieve this, immune cells are equipped with a plethora of mechanisms designed to recognise disruptions in homeostasis and to respond to these deviations. These in turn will be influenced by an individual’s genetic makeup and their history of antigen experience. We believe that antigen-specific immunotherapy, which aims at stimulating immune cells to target tumours, will be more successful if applied at a time when cancer cells are few within the body, helping to prevent relapse rather than to treat large tumours. Importantly, we believe that immunotherapy can be tilted towards a positive outcome if applied at a time when the patient’s entire wellbeing has first been taken into consideration and interventions have been taken to improve their mental health, gut microbiota, and approach to exercise prior to receiving therapy as well as during immunotherapeutic interventions.
In a time of “precision medicine” where most scientists believe that the treatment needs to be tailored to each individual patient, we would like to put forward the idea that preparing patients physically, psychologically, and microbiologically will improve the potency of any immunotherapy.

Author Contributions

Conceptualisation and writing, S.E.B.M., C.P.-S., J.R.D.P. and J.E.T.; visualisation, S.E.B.M. and C.P.-S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors thank Albert Puig-Saenz for helping with the visual representations of the graphical abstract and figure, which were created with BioRender.com (accessed on 18 July 2023).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Twomey, J.D.; Zhang, B. Cancer Immunotherapy Update: FDA-Approved Checkpoint Inhibitors and Companion Diagnostics. APPS J. 2021, 23, 39. [Google Scholar] [CrossRef]
  2. Conry, R.M.; Westbrook, B.; McKee, S.; Norwood, T.G. Talimogene Laherparepvec: First in Class Oncolytic Virotherapy. Hum. Vaccin. Immunother. 2018, 14, 839–846. [Google Scholar] [CrossRef] [Green Version]
  3. Ostrom, Q.T.; Cioffi, G.; Waite, K.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2014–2018. Neuro-Oncology 2021, 23, iii1–iii105. [Google Scholar] [CrossRef]
  4. Ohgaki, H. Epidemiology of Brain Tumors. In Cancer Epidemiology: Modifiable Factors; Verma, M., Ed.; Humana Press: Totowa, NJ, USA, 2009; pp. 323–342. ISBN 978-1-60327-492-0. [Google Scholar]
  5. Grossman, S.A.; Batara, J.F. Current Management of Glioblastoma Multiforme. Semin. Oncol. 2004, 31, 635–644. [Google Scholar] [CrossRef]
  6. Zhang, K.; Wang, X.; Zhou, B.; Zhang, L. The Prognostic Value of MGMT Promoter Methylation in Glioblastoma Multiforme: A Meta-Analysis. Fam. Cancer 2013, 12, 4492013458. [Google Scholar] [CrossRef]
  7. Malkki, H. Glioblastoma Vaccine Therapy Disappointment in Phase III Trial. Nat. Rev. Neurol. 2016, 12, 190. [Google Scholar] [CrossRef]
  8. Ravi, V.M.; Neidert, N.; Will, P.; Joseph, K.; Maier, J.P.; Kückelhaus, J.; Vollmer, L.; Goeldner, J.M.; Behringer, S.P.; Scherer, F.; et al. T-Cell Dysfunction in the Glioblastoma Microenvironment Is Mediated by Myeloid Cells Releasing Interleukin-10. Nat. Commun. 2022, 13, 925. [Google Scholar] [CrossRef] [PubMed]
  9. Woroniecka, K.I.; Rhodin, K.E.; Chongsathidkiet, P.; Keith, K.A.; Fecci, P.E. T-Cell Dysfunction in Glioblastoma: Applying a New Framework. Clin. Cancer Res. 2018, 24, 3792–3802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Mirzaei, R.; Sarkar, S.; Yong, V.W. T Cell Exhaustion in Glioblastoma: Intricacies of Immune Checkpoints. Trends Immunol. 2017, 38, 104–115. [Google Scholar] [CrossRef]
  11. Rudnick, J.D.; Fink, K.L.; Landolfi, J.C.; Markert, J.; Piccioni, D.E.; Glantz, M.J.; Swanson, S.J.; Gringeri, A.; Yu, J. Immunological Targeting of CD133 in Recurrent Glioblastoma: A Multi-Center Phase I Translational and Clinical Study of Autologous CD133 Dendritic Cell Immunotherapy. JCO 2017, 35, 2059. [Google Scholar] [CrossRef]
  12. Fadul, C.E.; Fisher, J.L.; Hampton, T.H.; Lallana, E.C.; Li, Z.; Gui, J.; Szczepiorkowski, Z.M.; Tosteson, T.D.; Rhodes, C.H.; Wishart, H.A.; et al. Immune Response in Patients with Newly Diagnosed Glioblastoma Multiforme Treated with Intranodal Autologous Tumor Lysate-Dendritic Cell Vaccination After Radiation Chemotherapy. J. Immunother. 2011, 34, 382. [Google Scholar] [CrossRef] [Green Version]
  13. Woroniecka, K.; Fecci, P.E. Immuno-Synergy? Neoantigen Vaccines and Checkpoint Blockade in Glioblastoma. Neuro-Oncology 2020, 22, 1233–1234. [Google Scholar] [CrossRef] [PubMed]
  14. Dohnal, A.M.; Witt, V.; Hügel, H.; Holter, W.; Gadner, H.; Felzmann, T. Phase I Study of Tumor Ag-Loaded IL-12 Secreting Semi-Mature DC for the Treatment of Pediatric Cancer. Cytotherapy 2007, 9, 755–770. [Google Scholar] [CrossRef]
  15. Chang, C.-N.; Huang, Y.-C.; Yang, D.-M.; Kikuta, K.; Wei, K.-J.; Kubota, T.; Yang, W.-K. A Phase I/II Clinical Trial Investigating the Adverse and Therapeutic Effects of a Postoperative Autologous Dendritic Cell Tumor Vaccine in Patients with Malignant Glioma. J. Clin. Neurosci. 2011, 18, 1048–1054. [Google Scholar] [CrossRef]
  16. Wen, P.Y.; Reardon, D.A.; Armstrong, T.S.; Phuphanich, S.; Aiken, R.D.; Landolfi, J.C.; Curry, W.T.; Zhu, J.-J.; Glantz, M.; Peereboom, D.M.; et al. A Randomized Double-Blind Placebo-Controlled Phase II Trial of Dendritic Cell Vaccine ICT-107 in Newly Diagnosed Patients with Glioblastoma. Clin. Cancer Res. 2019, 25, 5799–5807. [Google Scholar] [CrossRef] [PubMed]
  17. Inogés, S.; Tejada, S.; de Cerio, A.L.-D.; Gállego Pérez-Larraya, J.; Espinós, J.; Idoate, M.A.; Domínguez, P.D.; de Eulate, R.G.; Aristu, J.; Bendandi, M.; et al. A Phase II Trial of Autologous Dendritic Cell Vaccination and Radiochemotherapy Following Fluorescence-Guided Surgery in Newly Diagnosed Glioblastoma Patients. J. Transl. Med. 2017, 15, 104. [Google Scholar] [CrossRef] [Green Version]
  18. Wen, P.Y.; Reardon, D.A.; Forst, D.A.; Lee, E.Q.; Haas, B.; Daoud, T.; Berthoud, T.; Diaz-Mitoma, F.; Anderson, D.E.; Lassman, A.B.; et al. Evaluation of Tumor Responses and Overall Survival in Patients with Recurrent Glioblastoma (GBM) from a Phase IIa Trial of a CMV Vaccine Immunotherapeutic Candidate (VBI-1901). JCO 2022, 40, 2014. [Google Scholar] [CrossRef]
  19. Wang, Q.-T.; Nie, Y.; Sun, S.-N.; Lin, T.; Han, R.-J.; Jiang, J.; Li, Z.; Li, J.-Q.; Xiao, Y.-P.; Fan, Y.-Y.; et al. Tumor-Associated Antigen-Based Personalized Dendritic Cell Vaccine in Solid Tumor Patients. Cancer Immunol. Immunother. 2020, 69, 1375–1387. [Google Scholar] [CrossRef]
  20. Berneman, Z.N.; Anguille, S.; Willemen, Y.; de Velde, A.V.; Germonpre, P.; Huizing, M.; Van Tendeloo, V.; Saevels, K.; Rutsaert, L.; Vermeulen, K.; et al. Vaccination of Cancer Patients with Dendritic Cells Electroporated with MRNA Encoding the Wilms’ Tumor 1 Protein (WT1): Correlation of Clinical Effect and Overall Survival with T-Cell Response. Cytotherapy 2019, 21, S10. [Google Scholar] [CrossRef]
  21. Peereboom, D.M.; Nabors, L.B.; Kumthekar, P.; Badruddoja, M.A.; Fink, K.L.; Lieberman, F.S.; Phuphanich, S.; Dunbar, E.M.; Walbert, T.; Schiff, D.; et al. Phase 2 Trial of SL-701 in Relapsed/Refractory (r/r) Glioblastoma (GBM): Correlation of Immune Response with Longer-Term Survival. JCO 2018, 36, 2058. [Google Scholar] [CrossRef]
  22. Rahman, M.; Ghiaseddin, A.; Deleyrolle, P.; Peters, K.B.; Archer, G.; Sampson, J.; Mitchell, D. Phase II Randomized, Blinded, Placebo-Controlled Trial Testing Pp65 CMV MRNA Dendritic Cell Vaccine and Tetanus-Diphtheria Toxoid for Newly Diagnosed GBM (ATTAC II, NCT02465268). Neuro-Oncologyogy 2022, 24, vii60–vii61. [Google Scholar] [CrossRef]
  23. Dutoit, V.; Marinari, E.; Dietrich, P.-Y.; Migliorini, D. Combination of the IMA950/Poly-ICLC Multipeptide Vaccine with Pembrolizumab in Relapsing Glioblastoma Patients. Neuro Oncol. 2020, 22, ii34. [Google Scholar] [CrossRef]
  24. Sloan, A.E.; Dansey, R.; Zamorano, L.; Barger, G.; Hamm, C.; Diaz, F.; Baynes, R.; Wood, G. Adoptive Immunotherapy in Patients with Recurrent Malignant Glioma: Preliminary Results of Using Autologous Whole-Tumor Vaccine plus Granulocyte-Macrophage Colony-Stimulating Factor and Adoptive Transfer of Anti-CD3-Activated Lymphocytes. Neurosurg. Focus. 2000, 9, e9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Batich, K.A.; Mitchell, D.A.; Healy, P.; Herndon, J.E.; Sampson, J.H. Once, Twice, Three Times a Finding: Reproducibility of Dendritic Cell Vaccine Trials Targeting Cytomegalovirus in Glioblastoma. Clin. Cancer Res. 2020, 26, 5297–5303. [Google Scholar] [CrossRef]
  26. Reardon, D.A.; Idbaih, A.; Vieito, M.; Tabatabai, G.; Stradella, A.; Ghiringhelli, F.; Burger, M.C.; Mildenberger, I.; González, M.; Hervieu, A.; et al. EO2401 Therapeutic Vaccine for Patients with Recurrent Glioblastoma: Phase 1/2 ROSALIE Study (NCT04116658). Neuro-Oncology 2022, 24, vii63. [Google Scholar] [CrossRef]
  27. Tarakanovskaya, M.G.; Chinburen, J.; Batchuluun, P.; Munkhzaya, C.; Purevsuren, G.; Dandii, D.; Hulan, T.; Oyungerel, D.; Kutsyna, G.A.; Reid, A.A.; et al. Open-Label Phase II Clinical Trial in 75 Patients with Advanced Hepatocellular Carcinoma Receiving Daily Dose of Tableted Liver Cancer Vaccine, Hepcortespenlisimut-L. J. Hepatocell. Carcinoma 2017, 4, 59–69. [Google Scholar] [CrossRef] [Green Version]
  28. Yao, Y.; Luo, F.; Tang, C.; Chen, D.; Qin, Z.; Hua, W.; Xu, M.; Zhong, P.; Yu, S.; Chen, D.; et al. Molecular Subgroups and B7-H4 Expression Levels Predict Responses to Dendritic Cell Vaccines in Glioblastoma: An Exploratory Randomized Phase II Clinical Trial. Cancer Immunol. Immunother. 2018, 67, 1777–1788. [Google Scholar] [CrossRef]
  29. Hu, J.L.; Omofoye, O.A.; Rudnick, J.D.; Kim, S.; Tighiouart, M.; Phuphanich, S.; Wang, H.; Mazer, M.; Ganaway, T.; Chu, R.M.; et al. A Phase I Study of Autologous Dendritic Cell Vaccine Pulsed with Allogeneic Stem-like Cell Line Lysate in Patients with Newly Diagnosed or Recurrent Glioblastoma. Clin. Cancer Res. 2022, 28, 689–696. [Google Scholar] [CrossRef]
  30. Parney, I.F.; Anderson, S.K.; Gustafson, M.P.; Steinmetz, S.; Peterson, T.E.; Kroneman, T.N.; Raghunathan, A.; O’Neill, B.P.; Buckner, J.C.; Solseth, M.; et al. Phase I Trial of Adjuvant Mature Autologous Dendritic Cell/Allogeneic Tumor Lysate Vaccines in Combination with Temozolomide in Newly Diagnosed Glioblastoma. Neuro-Oncology Adv. 2022, 4, vdac089. [Google Scholar] [CrossRef]
  31. Batich, K.A.; Reap, E.A.; Archer, G.E.; Sanchez-Perez, L.; Nair, S.K.; Schmittling, R.J.; Norberg, P.; Xie, W.; Herndon, J.E., II; Healy, P.; et al. Long-Term Survival in Glioblastoma with Cytomegalovirus Pp65-Targeted Vaccination. Clin. Cancer Res. 2017, 23, 1898–1909. [Google Scholar] [CrossRef] [Green Version]
  32. Ahluwalia, M.S.; Reardon, D.A.; Abad, A.P.; Curry, W.T.; Wong, E.T.; Figel, S.A.; Mechtler, L.L.; Peereboom, D.M.; Hutson, A.D.; Withers, H.G.; et al. Phase IIa Study of SurVaxM Plus Adjuvant Temozolomide for Newly Diagnosed Glioblastoma. J. Clin. Oncol. 2023, 41, 1453–1465. [Google Scholar] [CrossRef] [PubMed]
  33. Wick, W.; Dietrich, P.-Y.; Kuttruff, S.; Hilf, N.; Frenzel, K.; Admon, A.; van der Burg, S.H.; von Deimling, A.; Gouttefangeas, C.; Kroep, J.R.; et al. GAPVAC-101: First-in-Human Trial of a Highly Personalized Peptide Vaccination Approach for Patients with Newly Diagnosed Glioblastoma. JCO 2018, 36, 2000. [Google Scholar] [CrossRef]
  34. Kodysh, J.; Bozkus, C.C.; Saxena, M.; Meseck, M.; Rubinsteyn, A.; O’Donnell, T.; Thin, T.H.; Brody, R.; Mandeli, J.; Bhardwaj, N.; et al. Phase I Study of Safety and Activity of Personalized Neoantigen-Based Vaccines in Combination with Tumor Treating Fields for Newly Diagnosed Glioblastoma Patients. J. Immunother. Cancer 2021, 9, 334. [Google Scholar] [CrossRef]
  35. Wheeler, L.A.; Manzanera, A.G.; Bell, S.D.; Cavaliere, R.; McGregor, J.M.; Grecula, J.C.; Newton, H.B.; Lo, S.S.; Badie, B.; Portnow, J.; et al. Phase II Multicenter Study of Gene-Mediated Cytotoxic Immunotherapy as Adjuvant to Surgical Resection for Newly Diagnosed Malignant Glioma. Neuro-Oncology 2016, 18, 1137–1145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Jouanneau, E.; Black, K.L.; Veiga, L.; Cordner, R.; Goverdhana, S.; Zhai, Y.; Zhang, X.; Panwar, A.; Mardiros, A.; Wang, H.; et al. Intrinsically De-Sialylated CD103+ CD8 T Cells Mediate Beneficial Anti-Glioma Immune Responses. Cancer Immunol. Immunother. 2014, 63, 911–924. [Google Scholar] [CrossRef]
  37. Chiocca, E.A.; Aguilar, L.K.; Bell, S.D.; Kaur, B.; Hardcastle, J.; Cavaliere, R.; McGregor, J.; Lo, S.; Ray-Chaudhuri, A.; Chakravarti, A.; et al. Phase IB Study of Gene-Mediated Cytotoxic Immunotherapy Adjuvant to up-Front Surgery and Intensive Timing Radiation for Malignant Glioma. J. Clin. Oncol. 2011, 29, 3611–3619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Moertel, C.; Pluhar, G.E.; Olin, M. Use of a Pan-Peptide Checkpoint Inhibitor in the Treatment of Central Nervous System Tumors. Neuro Oncol. 2023, 25, i81. [Google Scholar] [CrossRef]
  39. Zakharia, Y.; Johnson, T.S.; Colman, H.; Vahanian, N.N.; Link, C.J.; Kennedy, E.; Sadek, R.F.; Kong, F.M.; Vender, J.; Munn, D.; et al. A Phase I/II Study of the Combination of Indoximod and Temozolomide for Adult Patients with Temozolomide-Refractory Primary Malignant Brain Tumors. JCO 2014, 32, TPS2107. [Google Scholar] [CrossRef]
  40. Vik-Mo, E.O.; Nyakas, M.; Mikkelsen, B.V.; Moe, M.C.; Due-Tønnesen, P.; Suso, E.M.I.; Sæbøe-Larssen, S.; Sandberg, C.; Brinchmann, J.E.; Helseth, E.; et al. Therapeutic Vaccination against Autologous Cancer Stem Cells with MRNA-Transfected Dendritic Cells in Patients with Glioblastoma. Cancer Immunol. Immunother. 2013, 62, 1499–1509. [Google Scholar] [CrossRef] [Green Version]
  41. Bankiewicz, K.; Achrol, A.; Aghi, M.; Bexon, M.; Brenner, A.; Butowski, N.; Elder, B.; Floyd, J.; Lonser, R.; Merchant, F.; et al. MRI-Guided Convective Delivery of MDNA55, an Interleukin-4 Receptor Targeted Immunotherapy for the Treatment of Recurrent Glioblastoma. Neuro-Oncology 2017, 19, vi29. [Google Scholar] [CrossRef] [Green Version]
  42. Carpentier, A.F.; Verlut, C.; Ghiringhelli, F.; Bronnimann, C.; Ursu, R.; Fumet, J.D.; Gherga, E.; Lefort, F.; Belin, C.; Vernerey, D.; et al. Anti-Telomerase Vaccine in Patients with Newly Diagnosed, Unmethylated MGMT Glioblastoma: A Phase II Study. JCO 2023, 41, 2005. [Google Scholar] [CrossRef]
  43. Crittenden, M.; Bahjat, K.S.; Li, R.; Gore, P.; Fountain, C.; Hanson, B.; Skoble, J.; Lauer, P.; Murphy, A.L.; Dubensky, T.; et al. Phase I Study of Safety and Immunogenicity of ADU-623, a Live-Attenuated Listeria Monocytogenes Vaccine (ΔactA/ΔinlB) Expressing EGFRVIII and NY-ESO-1, in Patients with Who Grade III/IV Astrocytomas. J. Immunother. Cancer 2015, 3, P162. [Google Scholar] [CrossRef] [Green Version]
  44. Wick, W.; Wick, A.; Nowosielski, M.; Sahm, F.; Riehl, D.; Arzt, M.; von Deimling, A.; Bendszus, M.; Kickingereder, P.; Bonekamp, D.; et al. VXM01 Phase I Study in Patients with Resectable Progression of a Glioblastoma. JCO 2017, 35, 2061. [Google Scholar] [CrossRef]
  45. Wick, W.; Wick, A.; Chinot, O.L.; Van Den Bent, M.J.; De Vos, F.Y.F.L.; Mansour, M.; Podola, L.; Lubenau, H.; Platten, M. Oral DNA Vaccination Targeting VEGFR2 Combined with Anti-PDL1 Avelumab in Patients with Progressive Glioblastoma: Safety Run-in Results—NCT03750071. JCO 2020, 38, 3001. [Google Scholar] [CrossRef]
  46. Liau, L.M.; Ashkan, K.; Brem, S.; Campian, J.L.; Trusheim, J.E.; Iwamoto, F.M.; Tran, D.D.; Ansstas, G.; Cobbs, C.S.; Heth, J.A.; et al. Association of Autologous Tumor Lysate-Loaded Dendritic Cell Vaccination With Extension of Survival Among Patients With Newly Diagnosed and Recurrent Glioblastoma: A Phase 3 Prospective Externally Controlled Cohort Trial. JAMA Oncol. 2023, 9, 112–121. [Google Scholar] [CrossRef] [PubMed]
  47. Reardon, D.A.; Schuster, J.; Tran, D.D.; Fink, K.L.; Nabors, L.B.; Li, G.; Bota, D.A.; Lukas, R.V.; Desjardins, A.; Ashby, L.S.; et al. ReACT: Overall Survival from a Randomized Phase II Study of Rindopepimut (CDX-110) plus Bevacizumab in Relapsed Glioblastoma. JCO 2015, 33, 2009. [Google Scholar] [CrossRef]
  48. De Groot, J.F.; Cloughesy, T.F.; Pitz, M.W.; Narita, Y.; Nonomura, T. A Randomized, Multicenter Phase 2 Study of DSP-7888 Dosing Emulsion in Combination with Bevacizumab (Bev) versus Bev Alone in Patients with Recurrent or Progressive Glioblastoma. JCO 2018, 36, TPS2071. [Google Scholar] [CrossRef]
  49. Bloch, O.; Raizer, J.J.; Lim, M.; Sughrue, M.; Komotar, R.; Abrahams, J.; O’Rourke, D.; D’Ambrosio, A.; Bruce, J.N.; Parsa, A. Newly Diagnosed Glioblastoma Patients Treated with an Autologous Heat Shock Protein Peptide Vaccine: PD-L1 Expression and Response to Therapy. JCO 2015, 33, 2011. [Google Scholar] [CrossRef]
  50. Plautz, G.E.; Barnett, G.H.; Miller, D.W.; Cohen, B.H.; Prayson, R.A.; Krauss, J.C.; Luciano, M.; Kangisser, D.B.; Shu, S. Systemic T Cell Adoptive Immunotherapy of Malignant Gliomas. J. Neurosurg. 1998, 89, 42–51. [Google Scholar] [CrossRef] [Green Version]
  51. Sampson, J.H.; Schmittling, R.J.; Archer, G.E.; Congdon, K.L.; Nair, S.K.; Reap, E.A.; Desjardins, A.; Friedman, A.H.; Friedman, H.S.; Ii, J.E.H.; et al. A Pilot Study of IL-2Rα Blockade during Lymphopenia Depletes Regulatory T-Cells and Correlates with Enhanced Immunity in Patients with Glioblastoma. PLoS ONE 2012, 7, e31046. [Google Scholar] [CrossRef]
  52. Vlahovic, G.; Archer, G.E.; Reap, E.; Desjardins, A.; Peters, K.B.; Randazzo, D.; Healy, P.; Herndon, J.E.; Friedman, A.H.; Friedman, H.S.; et al. Phase I Trial of Combination of Antitumor Immunotherapy Targeted against Cytomegalovirus (CMV) plus Regulatory T-Cell Inhibition in Patients with Newly-Diagnosed Glioblastoma Multiforme (GBM). JCO 2016, 34, e13518. [Google Scholar] [CrossRef]
  53. Schuster, J.; Lai, R.K.; Recht, L.D.; Reardon, D.A.; Paleologos, N.A.; Groves, M.D.; Mrugala, M.M.; Jensen, R.; Baehring, J.M.; Sloan, A.; et al. A Phase II, Multicenter Trial of Rindopepimut (CDX-110) in Newly Diagnosed Glioblastoma: The ACT III Study. Neuro-Oncology 2015, 17, 854–861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Fenstermaker, R.A.; Ciesielski, M.J.; Qiu, J.; Yang, N.; Frank, C.L.; Lee, K.P.; Mechtler, L.R.; Belal, A.; Ahluwalia, M.S.; Hutson, A.D. Clinical Study of a Survivin Long Peptide Vaccine (SurVaxM) in Patients with Recurrent Malignant Glioma. Cancer Immunol. Immunother. 2016, 65, 1339–1352. [Google Scholar] [CrossRef] [Green Version]
  55. Bota, D.A.; Taylor, T.H.; Piccioni, D.E.; Duma, C.M.; LaRocca, R.V.; Kesari, S.; Carrillo, J.A.; Abedi, M.; Aiken, R.D.; Hsu, F.P.K.; et al. Phase 2 Study of AV-GBM-1 (a Tumor-Initiating Cell Targeted Dendritic Cell Vaccine) in Newly Diagnosed Glioblastoma Patients: Safety and Efficacy Assessment. J. Exp. Clin. Cancer Res. 2022, 41, 344. [Google Scholar] [CrossRef] [PubMed]
  56. Weller, M.; Butowski, N.; Tran, D.D.; Recht, L.D.; Lim, M.; Hirte, H.; Ashby, L.; Mechtler, L.; Goldlust, S.A.; Iwamoto, F.; et al. Rindopepimut with Temozolomide for Patients with Newly Diagnosed, EGFRvIII-Expressing Glioblastoma (ACT IV): A Randomised, Double-Blind, International Phase 3 Trial. Lancet Oncol. 2017, 18, 1373–1385. [Google Scholar] [CrossRef] [Green Version]
  57. Desjardins, A.; Sampson, J.H.; Peters, K.B.; Vlahovic, G.; Randazzo, D.; Threatt, S.; Herndon, J.E.; Boulton, S.; Lally-Goss, D.; McSherry, F.; et al. Patient Survival on the Dose Escalation Phase of the Oncolytic Polio/Rhinovirus Recombinant (PVSRIPO) against WHO Grade IV Malignant Glioma (MG) Clinical Trial Compared to Historical Controls. JCO 2016, 34, 2061. [Google Scholar] [CrossRef]
  58. Spira, A.; Hansen, A.R.; Harb, W.A.; Curtis, K.K.; Koga-Yamakawa, E.; Origuchi, M.; Li, Z.; Ertik, B.; Shaib, W.L. Multicenter, Open-Label, Phase I Study of DSP-7888 Dosing Emulsion in Patients with Advanced Malignancies. Targ. Oncol. 2021, 16, 461–469. [Google Scholar] [CrossRef]
  59. Migliorini, D.; Dutoit, V.; Allard, M.; Grandjean Hallez, N.; Marinari, E.; Widmer, V.; Philippin, G.; Corlazzoli, F.; Gustave, R.; Kreutzfeldt, M.; et al. Phase I/II Trial Testing Safety and Immunogenicity of the Multipeptide IMA950/Poly-ICLC Vaccine in Newly Diagnosed Adult Malignant Astrocytoma Patients. Neuro-Oncology 2019, 21, 923–933. [Google Scholar] [CrossRef] [Green Version]
  60. Prins, R.M.; Wang, X.; Soto, H.; Young, E.; Lisiero, D.N.; Fong, B.; Everson, R.; Yong, W.H.; Lai, A.; Li, G.; et al. Comparison of Glioma-Associated Antigen Peptide-Loaded Versus Autologous Tumor Lysate-Loaded Dendritic Cell Vaccination in Malignant Glioma Patients. J. Immunother. 2013, 36, 152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Liau, L.M.; Prins, R.M.; Kiertscher, S.M.; Odesa, S.K.; Kremen, T.J.; Giovannone, A.J.; Lin, J.-W.; Chute, D.J.; Mischel, P.S.; Cloughesy, T.F.; et al. Dendritic Cell Vaccination in Glioblastoma Patients Induces Systemic and Intracranial T-Cell Responses Modulated by the Local Central Nervous System Tumor Microenvironment. Clin. Cancer Res. 2005, 11, 5515–5525. [Google Scholar] [CrossRef] [Green Version]
  62. Bloch, O.; Crane, C.A.; Fuks, Y.; Kaur, R.; Aghi, M.K.; Berger, M.S.; Butowski, N.A.; Chang, S.M.; Clarke, J.L.; McDermott, M.W.; et al. Heat-Shock Protein Peptide Complex–96 Vaccination for Recurrent Glioblastoma: A Phase II, Single-Arm Trial. Neuro-Oncology 2014, 16, 274–279. [Google Scholar] [CrossRef] [Green Version]
  63. Thompson, E.M.; Landi, D.; Brown, M.C.; Friedman, H.S.; McLendon, R.; Herndon, J.E.; Buckley, E.; Bolognesi, D.P.; Lipp, E.; Schroeder, K.; et al. Recombinant Polio-Rhinovirus Immunotherapy for Recurrent Paediatric High-Grade Glioma: A Phase 1b Trial. Lancet Child. Adolesc. Health 2023, 7, 471–478. [Google Scholar] [CrossRef]
  64. Fu, S.; Piccioni, D.E.; Liu, H.; Lukas, R.V.; Aregawi, D.; Yamaguchi, K.; Whicher, K.; Chen, Y.-L.; Poola, N.; Eddy, J.; et al. Initial Phase 1 Study of WT2725 Dosing Emulsion in Patients with Advanced Malignancies. JCO 2017, 35, 2066. [Google Scholar] [CrossRef]
  65. Dees, K.J.; Koo, H.; Humphreys, J.F.; Hakim, J.A.; Crossman, D.K.; Crowley, M.R.; Nabors, L.B.; Benveniste, E.N.; Morrow, C.D.; McFarland, B.C. Human Gut Microbial Communities Dictate Efficacy of Anti-PD-1 Therapy in a Humanized Microbiome Mouse Model of Glioma. Neuro-Oncol. Adv. 2021, 3, vdab023. [Google Scholar] [CrossRef] [PubMed]
  66. Martínez-Garay, C.; Djouder, N. Dietary Interventions and Precision Nutrition in Cancer Therapy. Trends Mol. Med. 2023, 29, 489–511. [Google Scholar] [CrossRef]
  67. Lussier, D.M.; Woolf, E.C.; Johnson, J.L.; Brooks, K.S.; Blattman, J.N.; Scheck, A.C. Enhanced Immunity in a Mouse Model of Malignant Glioma Is Mediated by a Therapeutic Ketogenic Diet. BMC Cancer 2016, 16, 310. [Google Scholar] [CrossRef] [Green Version]
  68. Hansen, A.; Søgaard, K.; Minet, L.R. Development of an Exercise Intervention as Part of Rehabilitation in a Glioblastoma Multiforme Survivor during Irradiation Treatment: A Case Report. Disabil. Rehabil. 2019, 41, 1608–1614. [Google Scholar] [CrossRef] [Green Version]
  69. Huang, C.-W.; Chang, Y.-H.; Lee, H.-H.; Wu, J.-Y.; Huang, J.-X.; Chung, Y.-H.; Hsu, S.-T.; Chow, L.-P.; Wei, K.-C.; Huang, F.-T. Irisin, an Exercise Myokine, Potently Suppresses Tumor Proliferation, Invasion, and Growth in Glioma. FASEB J. 2020, 34, 9678–9693. [Google Scholar] [CrossRef]
  70. Conen, K.L.; Schüpbach, R.; Handschin, B.; Zwahlen, D.; Voss, M.; Eisele, G.; Rentsch, K.; Beyrau, R.; Vogt, D.R.; Katan, M.; et al. Prospective Evaluation of Stress in Patients with Newly Diagnosed Glioblastoma and in a Close Partner (TOGETHER-Study). JCO 2017, 35, e13524. [Google Scholar] [CrossRef]
  71. Stupp, R.; Hegi, M.E.; Mason, W.P.; van den Bent, M.J.; Taphoorn, M.J.B.; Janzer, R.C.; Ludwin, S.K.; Allgeier, A.; Fisher, B.; Belanger, K.; et al. Effects of Radiotherapy with Concomitant and Adjuvant Temozolomide versus Radiotherapy Alone on Survival in Glioblastoma in a Randomised Phase III Study: 5-Year Analysis of the EORTC-NCIC Trial. Lancet Oncol. 2009, 10, 459–466. [Google Scholar] [CrossRef] [PubMed]
  72. Keats, M.R.; Grandy, S.A.; Blanchard, C.; Fowles, J.R.; Neyedli, H.F.; Weeks, A.C.; MacNeil, M.V. The Impact of Resistance Exercise on Muscle Mass in Glioblastoma in Survivors (RESIST): Protocol for a Randomized Controlled Trial. JMIR Res. Protoc. 2022, 11, e37709. [Google Scholar] [CrossRef]
  73. Klein, P.; Tyrlikova, I.; Zuccoli, G.; Tyrlik, A.; Maroon, J.C. Treatment of Glioblastoma Multiforme with “Classic” 4:1 Ketogenic Diet Total Meal Replacement. Cancer Metab. 2020, 8, 24. [Google Scholar] [CrossRef] [PubMed]
  74. Yalamarty, S.S.K.; Filipczak, N.; Li, X.; Subhan, M.A.; Parveen, F.; Ataide, J.A.; Rajmalani, B.A.; Torchilin, V.P. Mechanisms of Resistance and Current Treatment Options for Glioblastoma Multiforme (GBM). Cancers 2023, 15, 2116. [Google Scholar] [CrossRef] [PubMed]
  75. Schreck, K.C.; Hsu, F.-C.; Berrington, A.; Henry-Barron, B.; Vizthum, D.; Blair, L.; Kossoff, E.H.; Easter, L.; Whitlow, C.T.; Barker, P.B.; et al. Feasibility and Biological Activity of a Ketogenic/Intermittent-Fasting Diet in Patients with Glioma. Neurology 2021, 97, e953–e963. [Google Scholar] [CrossRef] [PubMed]
  76. Martin-McGill, K.J.; Marson, A.G.; Tudur Smith, C.; Jenkinson, M.D. Ketogenic Diets as an Adjuvant Therapy in Glioblastoma (the KEATING Trial): Study Protocol for a Randomised Pilot Study. Pilot. Feasibility Stud. 2017, 3, 67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Anjum, K.; Shagufta, B.I.; Abbas, S.Q.; Patel, S.; Khan, I.; Shah, S.A.A.; Akhter, N.; Hassan, S.S.U. Current Status and Future Therapeutic Perspectives of Glioblastoma Multiforme (GBM) Therapy: A Review. Biomed. Pharm. 2017, 92, 681–689. [Google Scholar] [CrossRef]
  78. Rieger, J.; Bähr, O.; Maurer, G.D.; Hattingen, E.; Franz, K.; Brucker, D.; Walenta, S.; Kämmerer, U.; Coy, J.F.; Weller, M.; et al. ERGO: A Pilot Study of Ketogenic Diet in Recurrent Glioblastoma. Int. J. Oncol. 2014, 44, 1843–1852. [Google Scholar] [CrossRef] [Green Version]
  79. Schwartz, K.; Chang, H.T.; Nikolai, M.; Pernicone, J.; Rhee, S.; Olson, K.; Kurniali, P.C.; Hord, N.G.; Noel, M. Treatment of Glioma Patients with Ketogenic Diets: Report of Two Cases Treated with an IRB-Approved Energy-Restricted Ketogenic Diet Protocol and Review of the Literature. Cancer Metab. 2015, 3, 3. [Google Scholar] [CrossRef] [Green Version]
  80. Dardis, C.; Renda, L.; Honea, N.; Smith, K.; Nakaji, P.; Ashby, L.S.; Scheck, A.C. ACTR-15. Therapeutic Ketogenic Diet (KD) with Radiation and Chemotherapy for Newly Diagnosed Glioblastoma—Preliminary Results from NCT02046187. Neuro Oncol. 2017, 19, vi4. [Google Scholar] [CrossRef]
  81. Gresham, G.; Amaral, L.; Lockshon, L.; Levin, D.; Rudnick, J.; Provisor, A.; Shiao, S.; Bhowmick, N.; Irwin, S.; Freedland, S.; et al. ACTR-15. Phase 1 Trial of a Ketogenic Diet in Patients Receiving Standard-of-Care Treatment for Recently Diagnosed Glioblastoma. Neuro Oncol. 2019, 21, vi15. [Google Scholar] [CrossRef]
  82. Voss, M.; Wenger, K.J.; von Mettenheim, N.; Bojunga, J.; Vetter, M.; Diehl, B.; Franz, K.; Gerlach, R.; Ronellenfitsch, M.W.; Harter, P.N.; et al. Short-Term Fasting in Glioma Patients: Analysis of Diet Diaries and Metabolic Parameters of the ERGO2 Trial. Eur. J. Nutr. 2022, 61, 477–487. [Google Scholar] [CrossRef]
  83. Qayum, A.; Magotra, A.; Shah, S.M.; Nandi, U.; Sharma, P.R.; Shah, B.A.; Singh, S.K. Synergistic Combination of PMBA and 5-Fluorouracil (5-FU) in Targeting Mutant KRAS in 2D and 3D Colorectal Cancer Cells. Heliyon 2022, 8, e09103. [Google Scholar] [CrossRef]
  84. Brem, S.; Grossman, S.A.; Carson, K.A.; New, P.; Phuphanich, S.; Alavi, J.B.; Mikkelsen, T.; Fisher, J.D. New Approaches to Brain Tumor Therapy CNS Consortium Phase 2 Trial of Copper Depletion and Penicillamine as Antiangiogenesis Therapy of Glioblastoma. Neuro Oncol. 2005, 7, 246–253. [Google Scholar] [CrossRef]
  85. Cancer Research UK. Triple Negative Breast Cancer. Available online: https://www.cancerresearchuk.org/about-cancer/breast-cancer/types/triple-negative-breast-cancer (accessed on 22 January 2023).
  86. Breast Cancer Now. Triple Negative Breast Cancer. Available online: https://breastcancernow.org/information-support/facing-breast-cancer/diagnosed-breast-cancer/primary-breast-cancer/triple-negative-breast-cancer (accessed on 22 January 2023).
  87. Ghani, S.; Sochat, M.; Luo, J.; Tao, Y.; Ademuyiwa, F. Characteristics of Male Triple Negative Breast Cancer: A Population-Based Study. Breast J. 2020, 26, 1748–1755. [Google Scholar] [CrossRef] [PubMed]
  88. American Cancer Society. Triple-Negative Breast Cancer. Available online: https://www.cancer.org/cancer/types/breast-cancer/about/types-of-breast-cancer/triple-negative.html (accessed on 22 January 2023).
  89. Dent, R.; Trudeau, M.; Pritchard, K.I.; Hanna, W.M.; Kahn, H.K.; Sawka, C.A.; Lickley, L.A.; Rawlinson, E.; Sun, P.; Narod, S.A. Triple-Negative Breast Cancer: Clinical Features and Patterns of Recurrence. Clin. Cancer Res. 2007, 13, 4429–4434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Soares, R.F.; Garcia, A.R.; Monteiro, A.R.; Macedo, F.; Pereira, T.C.; Carvalho, J.C.; Pêgo, A.; Mariano, M.; Madeira, P.; Póvoa, S.; et al. Prognostic Factors for Early Relapse in Non-Metastatic Triple Negative Breast Cancer—Real World Data. Rep. Pract. Oncol. Radiother. 2021, 26, 563–572. [Google Scholar] [CrossRef] [PubMed]
  91. Allison, K.H.; Hammond, M.E.H.; Dowsett, M.; McKernin, S.E.; Carey, L.A.; Fitzgibbons, P.L.; Hayes, D.F.; Lakhani, S.R.; Chavez-MacGregor, M.; Perlmutter, J.; et al. Estrogen and Progesterone Receptor Testing in Breast Cancer: ASCO/CAP Guideline Update. J. Clin. Oncol. 2020, 38, 1346–1366. [Google Scholar] [CrossRef] [PubMed]
  92. Wang, J.; Xu, B. Targeted Therapeutic Options and Future Perspectives for HER2-Positive Breast Cancer. Sig. Transduct. Target. 2019, 4, 1–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Wahba, H.A.; El-Hadaad, H.A. Current Approaches in Treatment of Triple-Negative Breast Cancer. Cancer Biol. Med. 2015, 12, 106–116. [Google Scholar] [CrossRef] [PubMed]
  94. Biswas, T.; Efird, J.T.; Prasad, S.; Jindal, C.; Walker, P.R. The Survival Benefit of Neoadjuvant Chemotherapy and PCR among Patients with Advanced Stage Triple Negative Breast Cancer. Oncotarget 2017, 8, 112712–112719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Bianco, N.; Palazzo, A.; Pagan, E.; Bagnardi, V.; Milano, M.; De Maio, A.P.; Colleoni, M. Adjuvant Treatment for Triple Negative Breast Cancer with Residual Tumor after Neo-Adjuvant Chemotherapy. A Single Institutional Retrospective Analysis. Breast 2021, 59, 351–357. [Google Scholar] [CrossRef] [PubMed]
  96. Li, Y.; Zhang, H.; Merkher, Y.; Chen, L.; Liu, N.; Leonov, S.; Chen, Y. Recent Advances in Therapeutic Strategies for Triple-Negative Breast Cancer. J. Hematol. Oncol. 2022, 15, 121. [Google Scholar] [CrossRef]
  97. Tuohy, V.K.; Jaini, R.; Johnson, J.M.; Loya, M.G.; Wilk, D.; Downs-Kelly, E.; Mazumder, S. Targeted Vaccination against Human α-Lactalbumin for Immunotherapy and Primary Immunoprevention of Triple Negative Breast Cancer. Cancers 2016, 8, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Landry, I.; Sumbly, V.; Vest, M. Advancements in the Treatment of Triple-Negative Breast Cancer: A Narrative Review of the Literature. Cureus 2022, 14, e21970. [Google Scholar] [CrossRef]
  99. Gandhi, S.; Forsyth, P.; Opyrchal, M.; Ahmed, K.; Khong, H.; Attwood, K.; Levine, E.; O’Connor, T.; Early, A.; Fenstermaker, R.; et al. Phase IIa Study of Alpha-DC1 Vaccine against HER2/HER3, Chemokine Modulation Regimen and Pembrolizumab in Patients with Asymptomatic Brain Metastasis from Triple Negative or HER2+ Breast Cancer. J. Immunother. Cancer 2020, 8, 320. [Google Scholar] [CrossRef]
  100. Makhoul, I.; Ibrahim, S.M.; Abu-Rmaileh, M.; Jousheghany, F.; Siegel, E.R.; Rogers, L.J.; Lee, J.J.; Pina-Oviedo, S.; Post, G.R.; Beck, J.T.; et al. P10s-PADRE Vaccine Combined with Neoadjuvant Chemotherapy in ER-Positive Breast Cancer Patients Induces Humoral and Cellular Immune Responses. Oncotarget 2021, 12, 2252–2265. [Google Scholar] [CrossRef] [PubMed]
  101. Isakoff, S.J.; Tung, N.M.; Yin, J.; Tayob, N.; Parker, J.; Rosenberg, J.; Bardia, A.; Spring, L.; Park, H.; Collins, M.; et al. A Phase 1b Study of PVX-410 Vaccine in Combination with Pembrolizumab in Metastatic Triple Negative Breast Cancer (MTNBC). Cancer Res. 2022, 82, P2-14-17. [Google Scholar] [CrossRef]
  102. Isakoff, S.J.; Tolaney, S.M.; Tung, N.M.; Adams, S.; Soliman, H.H.; Brachtel, E.F.; Habin, K.R.; Bauer, L.J.; Ellisen, L.W.; Severgnini, M.; et al. A Phase 1b Study of Safety and Immune Response to PVX-410 Vaccine Alone and in Combination with Durvalumab (MEDI4736) in HLA-A2+ Patients Following Adjuvant Therapy for Stage 2/3 Triple Negative Breast Cancer. JCO 2017, 35, TPS1126. [Google Scholar] [CrossRef]
  103. Disis, M.; Liu, Y.; Stanton, S.; Gwin, W.; Coveler, A.; Liao, J.; Childs, J.; Cecil, D. A Phase I Dose Escalation Study of STEMVAC, a Multi-Antigen, Multi-Epitope Th1 Selective Plasmid-Based Vaccine, Targeting Stem Cell Associated Proteins in Patients with Advanced Breast Cancer. J. Immunother. Cancer 2022, 10, 546. [Google Scholar] [CrossRef]
  104. Kalli, K.R.; Block, M.S.; Kasi, P.M.; Erskine, C.L.; Hobday, T.J.; Dietz, A.; Padley, D.; Gustafson, M.P.; Shreeder, B.; Puglisi-Knutson, D.; et al. Folate Receptor Alpha Peptide Vaccine Generates Immunity in Breast and Ovarian Cancer Patients. Clin. Cancer Res. 2018, 24, 3014–3025. [Google Scholar] [CrossRef] [Green Version]
  105. Gao, T.; Cen, Q.; Lei, H. A Review on Development of MUC1-Based Cancer Vaccine. Biomed. Pharmacother. 2020, 132, 110888. [Google Scholar] [CrossRef] [PubMed]
  106. O’Shaughnessy, J.; Roberts, L.K.; Smith, J.L.; Levin, M.K.; Timis, R.; Finholt, J.P.; Burkeholder, S.B.; Tarnowski, J.; Muniz, L.S.; Melton, M.G.; et al. Safety and Initial Clinical Efficacy of a Dendritic Cell (DC) Vaccine in Locally Advanced, Triple-Negative Breast Cancer (TNBC) Patients (Pts). JCO 2016, 34, 1086. [Google Scholar] [CrossRef]
  107. Miao, L.; Zhang, Y.; Huang, L. MRNA Vaccine for Cancer Immunotherapy. Mol. Cancer 2021, 20, 41. [Google Scholar] [CrossRef] [PubMed]
  108. Rugo, H.S.; Chow, L.W.C.; Cortes, J.; Fasching, P.A.; Hsu, P.; Huang, C.-S.; Kim, S.-B.; Lu, Y.-S.; Melisko, M.E.; Nanda, R.; et al. Phase III, Randomized, Double-Blind, Placebo-Controlled Study to Evaluate the Efficacy and Safety of Adagloxad Simolenin (OBI-822) and OBI-821 Treatment in Patients with Early-Stage Triple-Negative Breast Cancer (TNBC) at High Risk for Recurrence. JCO 2020, 38, TPS599. [Google Scholar] [CrossRef]
  109. Jain, A.G.; Talati, C.; Pinilla-Ibarz, J. Galinpepimut-S (GPS): An Investigational Agent for the Treatment of Acute Myeloid Leukemia. Expert Opin. Investig. Drugs 2021, 30, 595–601. [Google Scholar] [CrossRef] [PubMed]
  110. Chung, V.; Kos, F.J.; Hardwick, N.; Yuan, Y.; Chao, J.; Li, D.; Waisman, J.; Li, M.; Zurcher, K.; Frankel, P.; et al. Evaluation of Safety and Efficacy of P53MVA Vaccine Combined with Pembrolizumab in Patients with Advanced Solid Cancers. Clin. Transl. Oncol. 2019, 21, 363–372. [Google Scholar] [CrossRef]
  111. Modi-1—Scancell. Available online: https://www.scancell.co.uk/modi-1 (accessed on 24 June 2023).
  112. Gheybi, E.; Salmanian, A.H.; Fooladi, A.A.I.; Salimian, J.; Hosseini, H.M.; Halabian, R.; Amani, J. Immunogenicity of Chimeric MUC1-HER2 Vaccine against Breast Cancer in Mice. Iran. J. Basic. Med. Sci. 2018, 21, 26–32. [Google Scholar] [CrossRef]
  113. Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. What Is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14. [Google Scholar] [CrossRef] [Green Version]
  114. Das, B.; Nair, G.B. Homeostasis and Dysbiosis of the Gut Microbiome in Health and Disease. J. Biosci. 2019, 44, 117. [Google Scholar] [CrossRef]
  115. Helmink, B.A.; Khan, M.A.W.; Hermann, A.; Gopalakrishnan, V.; Wargo, J.A. The Microbiome, Cancer, and Cancer Therapy. Nat. Med. 2019, 25, 377–388. [Google Scholar] [CrossRef]
  116. Lynn, D.J.; Benson, S.C.; Lynn, M.A.; Pulendran, B. Modulation of Immune Responses to Vaccination by the Microbiota: Implications and Potential Mechanisms. Nat. Rev. Immunol. 2022, 22, 33–46. [Google Scholar] [CrossRef] [PubMed]
  117. Fernández, M.F.; Reina-Pérez, I.; Astorga, J.M.; Rodríguez-Carrillo, A.; Plaza-Díaz, J.; Fontana, L. Breast Cancer and Its Relationship with the Microbiota. Int. J. Environ. Res. Public. Health 2018, 15, 1747. [Google Scholar] [CrossRef] [Green Version]
  118. Sadrekarimi, H.; Gardanova, Z.R.; Bakhshesh, M.; Ebrahimzadeh, F.; Yaseri, A.F.; Thangavelu, L.; Hasanpoor, Z.; Zadeh, F.A.; Kahrizi, M.S. Emerging Role of Human Microbiome in Cancer Development and Response to Therapy: Special Focus on Intestinal Microflora. J. Transl. Med. 2022, 20, 301. [Google Scholar] [CrossRef]
  119. Goedert, J.J.; Jones, G.; Hua, X.; Xu, X.; Yu, G.; Flores, R.; Falk, R.T.; Gail, M.H.; Shi, J.; Ravel, J.; et al. Investigation of the Association Between the Fecal Microbiota and Breast Cancer in Postmenopausal Women: A Population-Based Case-Control Pilot Study. J. Natl. Cancer Inst. 2015, 107, djv147. [Google Scholar] [CrossRef]
  120. Byrd, D.A.; Vogtmann, E.; Wu, Z.; Han, Y.; Wan, Y.; Clegg-Lamptey, J.-N.; Yarney, J.; Wiafe-Addai, B.; Wiafe, S.; Awuah, B.; et al. Associations of Fecal Microbial Profiles with Breast Cancer and Nonmalignant Breast Disease in the Ghana Breast Health Study. Int. J. Cancer 2021, 148, 2712–2723. [Google Scholar] [CrossRef] [PubMed]
  121. Ma, Z.; Qu, M.; Wang, X. Analysis of Gut Microbiota in Patients with Breast Cancer and Benign Breast Lesions. Pol. J. Microbiol. 2022, 71, 217–226. [Google Scholar] [CrossRef] [PubMed]
  122. Travis, R.C.; Key, T.J. Oestrogen Exposure and Breast Cancer Risk. Breast Cancer Res. 2003, 5, 239–247. [Google Scholar] [CrossRef] [PubMed]
  123. Parida, S.; Sharma, D. The Microbiome-Estrogen Connection and Breast Cancer Risk. Cells 2019, 8, 1642. [Google Scholar] [CrossRef] [Green Version]
  124. Devoy, C.; Flores Bueso, Y.; Tangney, M. Understanding and Harnessing Triple-Negative Breast Cancer-Related Microbiota in Oncology. Front. Oncol. 2022, 12, 1020121. [Google Scholar] [CrossRef]
  125. Treeck, O.; Schüler-Toprak, S.; Ortmann, O. Estrogen Actions in Triple-Negative Breast Cancer. Cells 2020, 9, 2358. [Google Scholar] [CrossRef]
  126. Gilbert, J.A.; Blaser, M.J.; Caporaso, J.G.; Jansson, J.K.; Lynch, S.V.; Knight, R. Current Understanding of the Human Microbiome. Nat. Med. 2018, 24, 392–400. [Google Scholar] [CrossRef] [PubMed]
  127. Urbaniak, C.; Cummins, J.; Brackstone, M.; Macklaim, J.M.; Gloor, G.B.; Baban, C.K.; Scott, L.; O’Hanlon, D.M.; Burton, J.P.; Francis, K.P.; et al. Microbiota of Human Breast Tissue. Appl. Environ. Microbiol. 2014, 80, 3007–3014. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Tzeng, A.; Sangwan, N.; Jia, M.; Liu, C.-C.; Keslar, K.S.; Downs-Kelly, E.; Fairchild, R.L.; Al-Hilli, Z.; Grobmyer, S.R.; Eng, C. Human Breast Microbiome Correlates with Prognostic Features and Immunological Signatures in Breast Cancer. Genome Med. 2021, 13, 60. [Google Scholar] [CrossRef]
  129. Banerjee, S.; Wei, Z.; Tan, F.; Peck, K.N.; Shih, N.; Feldman, M.; Rebbeck, T.R.; Alwine, J.C.; Robertson, E.S. Distinct Microbiological Signatures Associated with Triple Negative Breast Cancer. Sci. Rep. 2015, 5, 15162. [Google Scholar] [CrossRef] [Green Version]
  130. Banerjee, S.; Tian, T.; Wei, Z.; Shih, N.; Feldman, M.D.; Peck, K.N.; DeMichele, A.M.; Alwine, J.C.; Robertson, E.S. Distinct Microbial Signatures Associated with Different Breast Cancer Types. Front. Microbiol. 2018, 9, 951. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Banerjee, S.; Wei, Z.; Tian, T.; Bose, D.; Shih, N.N.C.; Feldman, M.D.; Khoury, T.; De Michele, A.; Robertson, E.S. Prognostic Correlations with the Microbiome of Breast Cancer Subtypes. Cell. Death Dis. 2021, 12, 1–14. [Google Scholar] [CrossRef]
  132. Warburton, D.E.R.; Nicol, C.W.; Bredin, S.S.D. Health Benefits of Physical Activity: The Evidence. CMAJ 2006, 174, 801–809. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Nieman, D.C.; Wentz, L.M. The Compelling Link between Physical Activity and the Body’s Defense System. J. Sport. Health Sci. 2019, 8, 201–217. [Google Scholar] [CrossRef]
  134. Hojman, P.; Stagaard, R.; Adachi-Fernandez, E.; Deshmukh, A.S.; Mund, A.; Olsen, C.H.; Keller, L.; Pedersen, B.K.; Gehl, J. Exercise Suppresses Tumor Growth Independent of High Fat Food Intake and Associated Immune Dysfunction. Sci. Rep. 2022, 12, 5476. [Google Scholar] [CrossRef]
  135. Wennerberg, E.; Lhuillier, C.; Rybstein, M.D.; Dannenberg, K.; Rudqvist, N.-P.; Koelwyn, G.J.; Jones, L.W.; Demaria, S. Exercise Reduces Immune Suppression and Breast Cancer Progression in a Preclinical Model. Oncotarget 2020, 11, 452–461. [Google Scholar] [CrossRef] [Green Version]
  136. Tiwari, P.; Blank, A.; Cui, C.; Schoenfelt, K.Q.; Zhou, G.; Xu, Y.; Khramtsova, G.; Olopade, F.; Shah, A.M.; Khan, S.A.; et al. Metabolically Activated Adipose Tissue Macrophages Link Obesity to Triple-Negative Breast Cancer. J. Exp. Med. 2019, 216, 1345–1358. [Google Scholar] [CrossRef] [PubMed]
  137. Vulczak, A.; de Souza, A.O.; Ferrari, G.D.; Azzolini, A.E.C.S.; Pereira-da-Silva, G.; Alberici, L.C. Moderate Exercise Modulates Tumor Metabolism of Triple-Negative Breast Cancer. Cells 2020, 9, 628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Chen, X.; Lu, W.; Zheng, W.; Gu, K.; Matthews, C.E.; Chen, Z.; Zheng, Y.; Shu, X.O. Exercise after Diagnosis of Breast Cancer in Association with Survival. Cancer Prev. Res. 2011, 4, 1409–1418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Janni, W.; Rack, B.; Friedl, T.; Müller, V.; Lorenz, R.; Rezai, M.; Tesch, H.; Heinrich, G.; Andergassen, U.; Harbeck, N.; et al. Lifestyle Intervention and Effect on Disease-Free Survival in Early Breast Cancer Pts: Interim Analysis from the Randomized SUCCESS C Study. Cancer Res. 2019, 79, GS5-03. [Google Scholar] [CrossRef]
  140. Lohmann, A.E.; Soldera, S.V.; Pimentel, I.; Ribnikar, D.; Ennis, M.; Amir, E.; Goodwin, P.J. Association of Obesity with Breast Cancer Outcome in Relation to Cancer Subtypes: A Meta-Analysis. J. Natl. Cancer Inst. 2021, 113, 1465–1475. [Google Scholar] [CrossRef] [PubMed]
  141. Zimmer, P.; Schmidt, M.E.; Prentzell, M.T.; Berdel, B.; Wiskemann, J.; Kellner, K.H.; Debus, J.; Ulrich, C.; Opitz, C.A.; Steindorf, K. Resistance Exercise Reduces Kynurenine Pathway Metabolites in Breast Cancer Patients Undergoing Radiotherapy. Front. Oncol. 2019, 9, 962. [Google Scholar] [CrossRef]
  142. Heng, B.; Bilgin, A.A.; Lovejoy, D.B.; Tan, V.X.; Milioli, H.H.; Gluch, L.; Bustamante, S.; Sabaretnam, T.; Moscato, P.; Lim, C.K.; et al. Differential Kynurenine Pathway Metabolism in Highly Metastatic Aggressive Breast Cancer Subtypes: Beyond IDO1-Induced Immunosuppression. Breast Cancer Res. 2020, 22, 113. [Google Scholar] [CrossRef]
  143. Alizadeh, A.M.; Isanejad, A.; Sadighi, S.; Mardani, M.; Kalaghchi, B.; Hassan, Z.M. High-Intensity Interval Training Can Modulate the Systemic Inflammation and HSP70 in the Breast Cancer: A Randomized Control Trial. J. Cancer Res. Clin. Oncol. 2019, 145, 2583–2593. [Google Scholar] [CrossRef]
  144. Liubomirski, Y.; Lerrer, S.; Meshel, T.; Rubinstein-Achiasaf, L.; Morein, D.; Wiemann, S.; Körner, C.; Ben-Baruch, A. Tumor-Stroma-Inflammation Networks Promote Pro-Metastatic Chemokines and Aggressiveness Characteristics in Triple-Negative Breast Cancer. Front. Immunol. 2019, 10, 757. [Google Scholar] [CrossRef] [Green Version]
  145. Clinton, S.K.; Giovannucci, E.L.; Hursting, S.D. The World Cancer Research Fund/American Institute for Cancer Research Third Expert Report on Diet, Nutrition, Physical Activity, and Cancer: Impact and Future Directions. J. Nutr. 2020, 150, 663–671. [Google Scholar] [CrossRef]
  146. Aznab, M.; Shojae, S.; Sorkheh, A.G.; Pia, K.E.; Rezaei, M. The Survival of Patients with Triple Negative Breast Cancer Undergoing Chemotherapy Along with Lifestyle Change Interventions: Survival of TNBC Patients. Arch. Breast Cancer 2023, 10, 66–73. [Google Scholar] [CrossRef]
  147. Swisher, A.K.; Abraham, J.; Bonner, D.; Gilleland, D.; Hobbs, G.; Kurian, S.; Yanosik, M.A.; Vona-Davis, L. Exercise and Dietary Advice Intervention for Survivors of Triple-Negative Breast Cancer: Effects on Body Fat, Physical Function, Quality of Life, and Adipokine Profile. Support. Care Cancer 2015, 23, 2995–3003. [Google Scholar] [CrossRef] [Green Version]
  148. Liao, M.; Qin, R.; Huang, W.; Zhu, H.-P.; Peng, F.; Han, B.; Liu, B. Targeting Regulated Cell Death (RCD) with Small-Molecule Compounds in Triple-Negative Breast Cancer: A Revisited Perspective from Molecular Mechanisms to Targeted Therapies. J. Hematol. Oncol. 2022, 15, 44. [Google Scholar] [CrossRef] [PubMed]
  149. de Groot, S.; Lugtenberg, R.T.; Cohen, D.; Welters, M.J.P.; Ehsan, I.; Vreeswijk, M.P.G.; Smit, V.T.H.B.M.; de Graaf, H.; Heijns, J.B.; Portielje, J.E.A.; et al. Fasting Mimicking Diet as an Adjunct to Neoadjuvant Chemotherapy for Breast Cancer in the Multicentre Randomized Phase 2 DIRECT Trial. Nat. Commun. 2020, 11, 3083. [Google Scholar] [CrossRef] [PubMed]
  150. Fasching, P.A.; Hein, A.; Kolberg, H.-C.; Häberle, L.; Uhrig, S.; Rübner, M.; Belleville, E.; Hack, C.C.; Fehm, T.N.; Janni, W.; et al. Pembrolizumab in Combination with Nab-Paclitaxel for the Treatment of Patients with Early-Stage Triple-Negative Breast Cancer—A Single-Arm Phase II Trial (NeoImmunoboost, AGO-B-041). Eur. J. Cancer 2023, 184, 1–9. [Google Scholar] [CrossRef] [PubMed]
  151. Harsini, S.; Wilson, D.; Saprunoff, H.; Allan, H.; Gleave, M.; Goldenberg, L.; Chi, K.N.; Kim-Sing, C.; Tyldesley, S.; Bénard, F. Outcome of Patients with Biochemical Recurrence of Prostate Cancer after PSMA PET/CT-Directed Radiotherapy or Surgery without Systemic Therapy. Cancer Imaging 2023, 23, 27. [Google Scholar] [CrossRef]
  152. Leslie, S.W.; Soon-Sutton, T.L.; Sajjad, H.; Siref, L.E. Prostate Cancer. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  153. 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]
  154. Fay, E.K.; Graff, J.N. Immunotherapy in Prostate Cancer. Cancers 2020, 12, 1752. [Google Scholar] [CrossRef]
  155. Vitkin, N.; Nersesian, S.; Siemens, D.R.; Koti, M. The Tumor Immune Contexture of Prostate Cancer. Front. Immunol. 2019, 10, 603. [Google Scholar] [CrossRef] [Green Version]
  156. Fizazi, K.; Mella, P.G.; Castellano, D.; Minatta, J.N.; Kalebasty, A.R.; Shaffer, D.; Limón, J.C.V.; López, H.M.S.; Armstrong, A.J.; Horvath, L.; et al. Nivolumab plus Docetaxel in Patients with Chemotherapy-Naïve Metastatic Castration-Resistant Prostate Cancer: Results from the Phase II CheckMate 9KD Trial. Eur. J. Cancer 2022, 160, 61–71. [Google Scholar] [CrossRef]
  157. Fizazi, K.; Retz, M.; Petrylak, D.P.; Goh, J.C.; Perez-Gracia, J.; Lacombe, L.; Zschäbitz, S.; Burotto, M.; Mahammedi, H.; Gravis, G.; et al. Nivolumab plus Rucaparib for Metastatic Castration-Resistant Prostate Cancer: Results from the Phase 2 CheckMate 9KD Trial. J. Immunother. Cancer 2022, 10, e004761. [Google Scholar] [CrossRef]
  158. Linch, M.; Papai, Z.; Takacs, I.; Imedio, E.R.; Kühnle, M.-C.; Derhovanessian, E.; Vogler, I.; Renken, S.; Graham, P.; Sahin, U.; et al. A First-in-Human (FIH) Phase I/IIa Clinical Trial Assessing a Ribonucleic Acid Lipoplex (RNA-LPX) Encoding Shared Tumor Antigens for Immunotherapy of Prostate Cancer; Preliminary Analysis of PRO-MERIT. J. Immunother. Cancer 2021, 9, 421. [Google Scholar] [CrossRef]
  159. Lee, S.C.; Ma, J.S.Y.; Kim, M.S.; Laborda, E.; Choi, S.-H.; Hampton, E.N.; Yun, H.; Nunez, V.; Muldong, M.T.; Wu, C.N.; et al. A PSMA-Targeted Bispecific Antibody for Prostate Cancer Driven by a Small-Molecule Targeting Ligand. Sci. Adv. 2021, 7, eabi8193. [Google Scholar] [CrossRef] [PubMed]
  160. Aggarwal, R.; Trihy, L.; Romero, E.H.; Sam, S.L.; Rastogi, M.; Kouchkovsky, I.D.; Small, E.J.; Feng, F.; Kwon, D.; Friedlander, T.; et al. A Phase Ib Study of a Single Priming Dose of 177Lu-PSMA-617 Coupled with Pembrolizumab in Metastatic Castration Resistant Prostate Cancer (MCRPC). Ann. Oncol. 2022, 33, S1173. [Google Scholar] [CrossRef]
  161. Einstein, D.J.; Wei, X.X.; Werner, L.; Ye, H.; Calagua, C.; Bubley, G.; Balk, S.P. A Phase II Study of Nivolumab in Patients with High-Risk Biochemically Recurrent (BCR) Prostate Cancer (PCa). JCO 2019, 37, TPS341. [Google Scholar] [CrossRef]
  162. Terrisse, S.; Zitvogel, L.; Kroemer, G. Effects of the Intestinal Microbiota on Prostate Cancer Treatment by Androgen Deprivation Therapy. Microb. Cell 2022, 9, 202–206. [Google Scholar] [CrossRef]
  163. Fujita, K.; Matsushita, M.; De Velasco, M.A.; Hatano, K.; Minami, T.; Nonomura, N.; Uemura, H. The Gut-Prostate Axis: A New Perspective of Prostate Cancer Biology through the Gut Microbiome. Cancers 2023, 15, 1375. [Google Scholar] [CrossRef]
  164. Kure, A.; Tsukimi, T.; Ishii, C.; Aw, W.; Obana, N.; Nakato, G.; Hirayama, A.; Kawano, H.; China, T.; Shimizu, F.; et al. Gut Environment Changes Due to Androgen Deprivation Therapy in Patients with Prostate Cancer. Prostate Cancer Prostatic Dis. 2023, 26, 323–330. [Google Scholar] [CrossRef]
  165. Liu, Y.; Jiang, H. Compositional Differences of Gut Microbiome in Matched Hormone-Sensitive and Castration-Resistant Prostate Cancer. Transl. Urol. 2020, 9, 1937–1944. [Google Scholar] [CrossRef]
  166. Che, B.; Zhang, W.; Xu, S.; Yin, J.; He, J.; Huang, T.; Li, W.; Yu, Y.; Tang, K. Prostate Microbiota and Prostate Cancer: A New Trend in Treatment. Front. Oncol. 2021, 11, 805459. [Google Scholar] [CrossRef]
  167. Rawla, P. Epidemiology of Prostate Cancer. World J. Oncol. 2019, 10, 63–89. [Google Scholar] [CrossRef] [Green Version]
  168. Santa Mina, D.; Au, D.; Alibhai, S.M.H.; Jamnicky, L.; Faghani, N.; Hilton, W.J.; Stefanyk, L.E.; Ritvo, P.; Jones, J.; Elterman, D.; et al. A Pilot Randomized Trial of Conventional versus Advanced Pelvic Floor Exercises to Treat Urinary Incontinence after Radical Prostatectomy: A Study Protocol. BMC Urol. 2015, 15, 94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Tsang, D.S.; Jones, J.M.; Samadi, O.; Shah, S.; Mitsakakis, N.; Catton, C.N.; Jeon, W.; To, J.; Breunis, H.; Alibhai, S.M.H. Healthy Bones Study: Can a Prescription Coupled with Education Improve Bone Health for Patients Receiving Androgen Deprivation Therapy?—A before/after Study. Support. Care Cancer 2018, 26, 2861–2869. [Google Scholar] [CrossRef]
  170. Patel, D.I.; Gallegos, A.M.; Sheikh, B.; Vardeman, S.; Liss, M.A. A Randomized Controlled Trial of a Home-Based Exercise Program on Prognostic Biomarkers in Men with Prostate Cancer: A Study Protocol. Contemp. Clin. Trials Commun. 2020, 20, 100659. [Google Scholar] [CrossRef]
  171. Winters-Stone, K.M.; Dobek, J.C.; Bennett, J.A.; Dieckmann, N.F.; Maddalozzo, G.F.; Ryan, C.W.; Beer, T.M. Resistance Training Reduces Disability in Prostate Cancer Survivors on Androgen Deprivation Therapy: Evidence from a Randomized Controlled Trial. Arch. Phys. Med. Rehabil. 2015, 96, 7–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Pernar, C.H.; Fall, K.; Rider, J.R.; Markt, S.C.; Adami, H.-O.; Andersson, S.-O.; Valdimarsdottir, U.; Andrén, O.; Mucci, L.A. A Walking Intervention among Men with Prostate Cancer: A Pilot Study. Clin. Genitourin. Cancer 2017, 15, e1021–e1028. [Google Scholar] [CrossRef] [PubMed]
  173. Thorsen, L.; Nilsen, T.S.; Raastad, T.; Courneya, K.S.; Skovlund, E.; Fosså, S.D. A Randomized Controlled Trial on the Effectiveness of Strength Training on Clinical and Muscle Cellular Outcomes in Patients with Prostate Cancer during Androgen Deprivation Therapy: Rationale and Design. BMC Cancer 2012, 12, 123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Segal, R.J.; Reid, R.D.; Courneya, K.S.; Malone, S.C.; Parliament, M.B.; Scott, C.G.; Venner, P.M.; Quinney, H.A.; Jones, L.W.; Slovinec D’Angelo, M.E.; et al. Resistance Exercise in Men Receiving Androgen Deprivation Therapy for Prostate Cancer. JCO 2003, 21, 1653–1659. [Google Scholar] [CrossRef]
  175. Brady, L.; Hayes, B.; Sheill, G.; Baird, A.-M.; Guinan, E.; Stanfill, B.; Vlajnic, T.; Casey, O.; Murphy, V.; Greene, J.; et al. Platelet Cloaking of Circulating Tumour Cells in Patients with Metastatic Prostate Cancer: Results from ExPeCT, a Randomised Controlled Trial. PLoS ONE 2020, 15, e0243928. [Google Scholar] [CrossRef]
  176. Katz, R.; Ahmed, M.A.; Safadi, A.; Abu Nasra, W.; Visoki, A.; Huckim, M.; Elias, I.; Nuriel-Ohayon, M.; Neuman, H. Characterization of Fecal Microbiome in Biopsy Positive Prostate Cancer Patients. BJUI Compass 2021, 3, 55–61. [Google Scholar] [CrossRef]
  177. Li, J.K.M.; Wang, L.L.; Lau, B.S.Y.; Tse, R.T.H.; Cheng, C.K.L.; Leung, S.C.H.; Wong, C.Y.P.; Tsui, S.K.W.; Teoh, J.Y.C.; Chiu, P.K.F.; et al. Oral Antibiotics Perturbation on Gut Microbiota after Prostate Biopsy. Front. Cell. Infect. Microbiol. 2022, 12, 959903. [Google Scholar] [CrossRef] [PubMed]
  178. Zhong, W.; Wu, K.; Long, Z.; Zhou, X.; Zhong, C.; Wang, S.; Lai, H.; Guo, Y.; Lv, D.; Lu, J.; et al. Gut Dysbiosis Promotes Prostate Cancer Progression and Docetaxel Resistance via Activating NF-ΚB-IL6-STAT3 Axis. Microbiome 2022, 10, 94. [Google Scholar] [CrossRef] [PubMed]
  179. Chang, P.-Y.; Huang, W.-Y.; Lin, C.-L.; Huang, T.-C.; Wu, Y.-Y.; Chen, J.-H.; Kao, C.-H. Propranolol Reduces Cancer Risk: A Population-Based Cohort Study. Medicine 2015, 94, e1097. [Google Scholar] [CrossRef] [PubMed]
  180. Zhang, M.; Wang, Q.; Sun, X.; Yin, Q.; Chen, J.; Xu, L.; Xu, C. Β2 -Adrenergic Receptor Signaling Drives Prostate Cancer Progression by Targeting the Sonic Hedgehog-Gli1 Signaling Activation. Prostate 2020, 80, 1328–1340. [Google Scholar] [CrossRef] [PubMed]
  181. Grytli, H.H.; Fagerland, M.W.; Fosså, S.D.; Taskén, K.A. Association Between Use of β-Blockers and Prostate Cancer–Specific Survival: A Cohort Study of 3561 Prostate Cancer Patients with High-Risk or Metastatic Disease. Eur. Urol. 2014, 65, 635–641. [Google Scholar] [CrossRef] [PubMed]
  182. McCarty, R. Chapter 4—The Fight-or-Flight Response: A Cornerstone of Stress Research. In Stress: Concepts, Cognition, Emotion, and Behavior; Fink, G., Ed.; Academic Press: San Diego, CA, USA, 2016; pp. 33–37. ISBN 978-0-12-800951-2. [Google Scholar]
  183. Dai, S.; Mo, Y.; Wang, Y.; Xiang, B.; Liao, Q.; Zhou, M.; Li, X.; Li, Y.; Xiong, W.; Li, G.; et al. Chronic Stress Promotes Cancer Development. Front. Oncol. 2020, 10, 1492. [Google Scholar] [CrossRef]
  184. Mohammadpour, H.; MacDonald, C.R.; Qiao, G.; Chen, M.; Dong, B.; Hylander, B.L.; McCarthy, P.L.; Abrams, S.I.; Repasky, E.A. Β2 Adrenergic Receptor-Mediated Signaling Regulates the Immunosuppressive Potential of Myeloid-Derived Suppressor Cells. J. Clin. Invest. 2019, 129, 5537–5552. [Google Scholar] [CrossRef] [Green Version]
  185. Liu, H.; Yang, J.; Zhang, Y.; Han, J.; Yang, Y.; Zhao, Z.; Dai, X.; Wang, H.; Ding, X.; Liu, Y.; et al. Psychologic Stress Drives Progression of Malignant Tumors via DRD2/HIF1α Signaling. Cancer Res. 2021, 81, 5353–5365. [Google Scholar] [CrossRef]
  186. Cain, D.W.; Cidlowski, J.A. Immune Regulation by Glucocorticoids. Nat. Rev. Immunol. 2017, 17, 233–247. [Google Scholar] [CrossRef]
  187. Ludolph, P.; Kunzler, A.M.; Stoffers-Winterling, J.; Helmreich, I.; Lieb, K. Interventions to Promote Resilience in Cancer Patients. Dtsch. Arztebl. Int. 2019, 51–52, 865–872. [Google Scholar] [CrossRef]
  188. Contreras-Rodriguez, O.; Reales-Moreno, M.; Fernández-Barrès, S.; Cimpean, A.; Arnoriaga-Rodríguez, M.; Puig, J.; Biarnés, C.; Motger-Albertí, A.; Cano, M.; Fernández-Real, J.M. Consumption of Ultra-Processed Foods Is Associated with Depression, Mesocorticolimbic Volume, and Inflammation. J. Affect. Disord. 2023, 335, 340–348. [Google Scholar] [CrossRef] [PubMed]
  189. Mayneris-Perxachs, J.; Castells-Nobau, A.; Arnoriaga-Rodríguez, M.; Martin, M.; de la Vega-Correa, L.; Zapata, C.; Burokas, A.; Blasco, G.; Coll, C.; Escrichs, A.; et al. Microbiota Alterations in Proline Metabolism Impact Depression. Cell. Metab. 2022, 34, 681–701.e10. [Google Scholar] [CrossRef] [PubMed]
  190. Valles-Colomer, M.; Falony, G.; Darzi, Y.; Tigchelaar, E.F.; Wang, J.; Tito, R.Y.; Schiweck, C.; Kurilshikov, A.; Joossens, M.; Wijmenga, C.; et al. The Neuroactive Potential of the Human Gut Microbiota in Quality of Life and Depression. Nat. Microbiol. 2019, 4, 623–632. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mechanisms of immune modulation by physical and psychological factors. Abbreviations: SAM, sympathetic–adreno–medullar; HPA, hypothalamic–pituitary–adrenal; DCs, dendritic cells; MDSCs, myeloid-derived suppressor cells; Tregs, regulatory T cells; HIF1α, hypoxia-inducible factor-1α; CRP, C-reactive protein; TGF-β, transforming growth factor-β; LPS, lipopolysaccharide.
Figure 1. Mechanisms of immune modulation by physical and psychological factors. Abbreviations: SAM, sympathetic–adreno–medullar; HPA, hypothalamic–pituitary–adrenal; DCs, dendritic cells; MDSCs, myeloid-derived suppressor cells; Tregs, regulatory T cells; HIF1α, hypoxia-inducible factor-1α; CRP, C-reactive protein; TGF-β, transforming growth factor-β; LPS, lipopolysaccharide.
Biomedicines 11 02100 g001
Table 1. Immunotherapy trials for GBM.
Table 1. Immunotherapy trials for GBM.
ClinicalTrials.gov IDVaccinePhaseStageReference
NCT02049489ICT-121ICompleted[11]
NCT00323115aDCs 1 + RT 2 + TMZ 3IICompleted[12]
NCT04277221aDCs/tumour antigen + RT + TMZIIIUnknown[13]
NCT01213407Trivax + RT + TMZIICompleted[14]
NCT02772094aDCs/tumour antigen + TMZIIUnknown[15]
NCT01280552ICT-107IICompleted[16]
NCT01006044aDCsIICompleted[17]
NCT03382977VBI-1901I/IIActive, not recruiting[18]
NCT02864368PEP-CMV + TMZITerminated
NCT02146066DCVax-L + TMZE.A. 4Available
NCT04968366aDCs pulsed with multiple neoantigen peptidesIRecruiting
NCT02709616mRNA-pulsed DCs + RT + TMZICompleted[19]
NCT02808364mRNA-pulsed aDCsICompleted[19]
NCT02649582mRNA-pulsed DCs + RT + TMZI/IIRecruiting[20]
NCT02078648SL-701I/IICompleted[21]
NCT05685004TVI-Brain-1 + RT + TMZII/IIINot yet recruiting
NCT02510950Personalised peptide + Poly-ICLC + TMZITerminated
NCT02465268pp65-shLAMP DCIIActive, not recruiting[22]
NCT04801147aDCsI/IIRecruiting
NCT01902771DCs + tumour lysate + ImiquimodITerminated
NCT04002804aDCs + autologous tumour lysateITerminated
NCT03665545IMA950I/IIActive, not recruiting[23]
NCT04842513Multipeptide plus XS15 + RT + TMZIRecruiting
NCT01290692TVI-Brain-1IICompleted[24]
NCT02366728CMV pp65 -LAMP mRNA-pulsed aDCs + TMZ + BasiliximabIICompleted[25]
NCT04116658EO2401 + Nivolumab/Nivolumab + BevacizumabIb/IIaActive, not recruiting[26]
NCT03916757V-BoostIIUnknown[27]
NCT01567202aDCs + Autogeneic glioma stem-like cells (A2B5+) + RT + TMZ IIUnknown[28]
NCT02010606Allogenic GBM stem-like lysate-pulsed aDCsICompleted[29]
NCT00643097PEP-3-KLH + GM-CSF + TMZIICompleted
NCT04963413pp65-fLAMP RNA-loaded aDCs + GM-CSF + TMZIActive, not recruiting
NCT01957956Allogenic tumour lysate-pulsed aDCsEarly IActive, not recruiting[30]
NCT01808820aDCs + Allogenic tumour lysate + ImiquimodICompleted
NCT04015700GNOS-PV01 + INO-9012IActive, not recruiting
NCT00639639pp65-LAMP mRNA-loaded DCs + Ttd 5ICompleted[31]
NCT03360708Allogenic tumour lysate-pulsed aDCsEarly IActive, not recruiting
NCT01081223TVI-Brain-1 + IL-2I/IICompleted[24]
NCT02722512HSPPC-96 + RTITerminated
NCT02455557SurVaxMIIActive, not recruiting[32]
NCT03914768Genetically modified tumour cells/antigens-pulsed aDCSIUnknown
NCT03615404aDCs + CMV RNA + CM-CSF + TtdIRecruiting
NCT02149225APVAC + Poly-ICLC + GM-CSFICompleted[33]
NCT03223103Personalised vaccine + Poly-ICLCIActive, not recruiting[34]
NCT05743595Personalised neoantigen DNA + RetifanlimabINot yet recruiting
NCT03688178aDCs + CMV pp65 + TMZ + VarlilumabIIRecruiting
NCT04888611GSC-DCV + CamrelizumabIIRecruiting
NCT00890032aDCs + Autologous tumour mRNAICompleted
NCT00589875AdV-tk + ValacyclovirIICompleted[35]
NCT01403285IMA950 + Cyclophosphamide + GM-CSFITerminated
NCT03927222aDCs + pp65-LAMP CMV mRNA + GM-CSF + TtdIITerminated
NCT00576537aDCs + Allogenic tumour lysateIICompleted[36]
NCT04573140RNA-LPIRecruiting
NCT00751270GliAtakICompleted[37]
NCT03422094NeoVax + Nivolumab + IpilimumabITerminated
NCT04642937GBM6-AD + Hp1a8 + ImiquimodIActive, not recruiting[38]
NCT02052648Indoximod + TMZI/IICompleted[39]
NCT01222221IMA950 + TMZ + RTICompleted
NCT02529072DCs + NivolumabICompleted
NCT00846456CSC-mRNA transfected DCsICompleted[40]
NCT01814813MDNA-55IITerminated[41]
NCT02287428NeoVax + PembrolizumabIRecruiting
NCT03018288HSPCC-96 + Pembrolizumab + RT + TMZICompleted
NCT04201873aDCs + Pembrolizumab + Poly-ICLC IRecruiting
NCT04280848UCPvaxIIRecruiting[42]
NCT01967758ADU-623ICompleted[43]
NCT02820584aDCs + GSCICompleted
NCT04552886aDCsIRecruiting
NCT02718443VXM01ICompleted[44]
NCT04523688aDCsIIRecruiting
NCT01759810DCsIUnknown
NCT03750071VXM01 + AvelumabI/IIActive, not recruiting[45]
NCT03879512aDCs + Tumour lysate + Cyclophosphamide + Nivolumab + IpilimumabI/IIRecruiting
NCT03395587aDCs + SoC 6IIRecruiting
NCT00045968DCVax-LIIIActive, not recruiting[46]
NCT01498328CDX-110 + BevacizumabIICompleted[47]
NCT05698199ITI-1001INot yet recruiting
NCT03149003DSP-7888 + BevacizumabIIICompleted[48]
NCT00905060HSPPC-96 + TMZ + SurgeryIICompleted[49]
NCT00003185Autologous tumour cells + SargramostimIICompleted[50]
NCT00626015PEP3-KLH + Daclizumab + TMZICompleted[51]
NCT00626483DCs loaded with CMV pp65-LAMP mRNAICompleted[52]
NCT00458601CDX-110 + TMZ + GM-CSFIIComplete[53]
NCT05100641AV-GBM-1IIINot recruiting yet
NCT01250470SurVaxMIComplete[54]
NCT03400917AV-GBM-1IIActive, not recruiting[55]
NCT05163080SurVaxM + Montanide + SargramostimIIRecruiting
NCT01480479CDX-110 + TMZ + GM-CSFIIICompleted[56]
NCT00576641aDCs + Autologous tumour lysateICompleted[36]
NCT01204684Resiquimod + Poly-ICLCIIActive, not recruiting
NCT01491893PVSRIPOICompleted[57]
NCT05557240NPVAC1/2 + Poly-ICLCIRecruiting
NCT04388033aDCs + IL-12I/IIRecruiting
NCT05356312Personalised neoantigen vaccineE.A. 4Available
NCT05283109P30-EPS + HiltonolINot yet recruiting
NCT02498665DSP-7888ICompleted[58]
NCT02800486CetuximabIIRecruiting
NCT04978727SurVaxMIRecruiting
NCT01920191IMA950 + Poly-ICLC + TMZI/IICompleted[59]
NCT00612001aDCsICompleted[60]
NCT00068510aDCs loaded with tumour lysateICompleted[61]
NCT04214392CAR T-CellsIRecruiting
NCT00293423GP96ICompleted[62]
NCT01522820DEC-205/NY-ESO-1 Fusion Protein CDX-1401 + SirolimusICompleted
NCT04808245H3K27M peptide + ImiquimodIRecruiting
NCT00069940Telomerase: 540–548 peptide + GM-CSFICompleted
NCT03043391PVSRIPOIbActive, not recruiting[63]
NCT00014573Surgery + Paclitaxel + Cyclophosphamide + Filgrastim + Autologous tumour cells + Sargramostim + Cisplatin + Carmustine + IL-2 + Autologous bone marrow/PBMC transplantationIICompleted
NCT01621542WT2725ICompleted[64]
NCT00004024Autologous tumour cells + Muromonab-CD3 + GM-CSF + IL-2IICompleted
1 Autologous dendritic cells; 2 radiotherapy; 3 temozolomide; 4 early access; 5 tetanus toxoid; 6 standard of care.
Table 2. Trials assessing exercise, diet, and microbiome in GBM patients.
Table 2. Trials assessing exercise, diet, and microbiome in GBM patients.
ClinicalTrials.gov IDInterventionPhaseStageReference
NCT03390569ExerciseN/A 1Completed
NCT05015543Personal training programmeN/ARecruiting
NCT02129335Impact of exercise on stress N/ATerminated[70]
NCT05431348Impact of stress and exercise on chemoradiation outcomeN/ARecruiting[71]
NCT05116137Circuit-based resistance exerciseN/AEnrolling by invitation[72]
NCT03501134NovoTTF 2 deviceN/ACompleted
NCT01865162Ketogenic dietICompleted[73]
NCT05708352Ketogenic dietIINot yet recruiting[74]
NCT02286167Atkins-based dietN/ACompleted[75]
NCT02939378Ketogenic dietI/IIUnknown
NCT03075514Ketogenic dietN/ACompleted[76]
NCT02302235Ketogenic dietIICompleted[73]
NCT00508456Methionine-restricted dietITerminated
NCT04730869Metabolic therapy programmeN/ARecruiting[77]
NCT00575146Ketogenic dietICompleted[78]
NCT04691960Ketogenic diet + MetforminIIRecruiting
NCT01535911Metabolic nutritional therapyN/AActive, not recruiting[79]
NCT02046187Ketogenic dietI/IITerminated[80]
NCT03451799Ketogenic dietIActive, not recruiting[81]
NCT05183204Ketogenic diet + Metformin + PaxalisibIIRecruiting
NCT03160599Ketogenic dietN/AUnknown
NCT03278249Modified Atkins ketogenic dietN/AActive, not recruiting
NCT02768389Modified Atkins diet + BevacizumabEarly ICompleted
NCT01754350Calorie-restricted ketogenic diet + Transient fastingN/ACompleted[82]
NCT00243022Boswellia serrata extract + Vitamin B12IITerminated[83]
NCT05326334Chemoradiation + Chemotherapy + Microbiome evaluationN/ARecruiting
NCT00003751Penicillamine + Low copper dietIICompleted[84]
NCT03631823Chemotherapy and/or radiotherapy + Correlation between microbiome and prognosisN/AUnknown
1 Not applicable; 2 tumour treating fields.
Table 3. Immunotherapy trials for TNBC.
Table 3. Immunotherapy trials for TNBC.
ClinicalTrials.gov IDVaccinePhaseStageReference
NCT04674306α-lactalbumin + ZymosanEarly IRecruiting[97]
NCT04024800AE37 peptide + PembrolizumabIIActive, not recruiting[98]
NCT03199040Neoantigen DNA + DurvalumabIActive, not recruiting
NCT04348747HER2/HER3 DCs + PembrolizumabIIaRecruiting[99]
NCT02348320Polyepitope DNAICompleted
NCT02938442P10s-PADRE with MONTANIDE ISA 51 VG + Doxorubicin + Cyclophosphamide + Paclitaxel + SurgeryIICompleted[100]
NCT03362060PVX-410 + PembrolizumabIbActive, not recruiting[101]
NCT02826434PVX-410 + Durvalumab + Poly-ICLCIbActive, not recruiting[102]
NCT05455658STEMVAC + SargramostimIIRecruiting[103]
NCT03606967Personalised neoantigen peptide + Carboplatin + Gemcitabine + Nab-Paclitaxel + Durvalumab + Tremelimumab + Poly-ICLCIIRecruiting
NCT03387085N-803 + ETBX-011 + ETBX-051 + ETBX-061 + GI-4000 + GI-6207 + GI-6301 + HaNK + Avelumab + Bevacizumab + Aldoxorubicin + Capecitabine + Cisplatin + Cyclophosphamide + 5-Fluorouracil + Leucovorin + Nab-PaclitaxelIb/IIActive, not recruiting
NCT03012100Multi-epitope folate receptor alpha + Cyclophosphamide + GM-CSFIIActive, not recruiting[104]
NCT00986609MUC1 + Poly-ICLCEarly ICompleted[105]
NCT02593227Folate receptor alpha + Cyclophosphamide + GM-CSFIICompleted[104]
NCT05504707HER2-/HER3-primed DC1IRecruiting
NCT04105582Neoantigen-pulsed aDCs 1ICompleted
NCT02018458Cyclin B1/WT1/CEF-pulsed DCs + Doxorubicin + Cyclophosphamide + Paclitaxel + CarboplatinICompleted[106]
NCT02316457RNA for shared tumour associated antigens + RNA for tumour specific antigensIActive, not recruiting[107]
NCT03562637OBI-822 + OBI-821IIIRecruiting[108]
NCT04634747PVX-410 + Pembrolizumab + ChemotherapyIINot yet recruiting
NCT05269381Personalised neoantigen + Pembrolizumab + Cyclophosphamide + GM-CSFIRecruiting
NCT03761914Galinpepimut-S + PembrolizumabI/IIActive, not recruiting[109]
NCT02432963P53MVA + PembrolizumabIActive, not recruiting[110]
NCT05329532Modi-1/Modi-1v + PembrolizumabI/IIRecruiting[111]
NCT00640861MUC1 + HER2/neu + CpG + GM-CSF + IFAEarly ICompleted[112]
NCT04879888Peptide-pulsed aDCsICompleted
NCT05035407KK-LC-1 TCR + Aldesleukin + Cyclophosphamide + FludarabineIRecruiting
1 Autologous dendritic cells.
Table 4. Trials assessing exercise, diet, and microbiome in TNBC patients.
Table 4. Trials assessing exercise, diet, and microbiome in TNBC patients.
ClinicalTrials.gov IDInterventionPhaseStageReference
NCT01498536Aerobic exerciseN/A 1Completed[147]
NCT03733119Methionine-restricted diet + ONC201IITerminated[148]
NCT04248998Fasting-mimicking diet + MetforminIIActive, not recruiting[149]
NCT05763992Fasting-like approach + SoC 2IIRecruiting
NCT03186937Methionine-restricted dietIITerminated
NCT02348320Caloric restriction diet + SABR 3IIRecruiting
NCT04677816Vitamin D3 + SoCIIRecruiting
NCT05198843Icosapent ethyl + DasatinibIb/IIRecruiting
NCT05037825ICI 4 + Microbiome evaluationN/ARecruiting
NCT03586297SoC + Correlation between microbiome composition and pCR 5N/ARecruiting
NCT04638751Chemotherapy + Correlation between microbiome, PFS, 6 and OS 7N/ARecruiting
NCT05916755Pembrolizumab and/or chemotherapy + microbiome analysis to establish predictive biomarkersN/ARecruiting
NCT03289819Pembrolizumab + Nab-Paclitaxel + Epirubicin + Cyclophosphamide + Correlation between microbiome and clinical outcomeIICompleted[150]
1 Not applicable; 2 standard of care; 3 stereotactic ablative radiotherapy; 4 immune checkpoint inhibitors; 5 pathological complete response; 6 progression-free survival; 7 overall survival.
Table 5. Immunotherapy trials for PCa.
Table 5. Immunotherapy trials for PCa.
ClinicalTrials.gov IDVaccinePhaseStageReference
NCT00003871Fowlpox prostate specific antigenIICompleted[156]
NCT00374049MUC1 + Poly-ICLC + GM-CSFICompleted
NCT00122005GVAXI/IIUnknown
NCT03815942ChAdOx1-MVA 5T4 + NivolumabI/IIUnknown[157]
NCT02234921Cyclophosphamide + Dribble + Imiquimod + CervarixICompleted
NCT01867333Enzalutamide + PROSTVAC-F/TRICOM + PROSTVAC-V/TRICOMIICompleted[158]
NCT04914195Leuprolide acetateIIIRecruiting
NCT01420965Sipuleucel-T + CT-011 + CyclophosphamideIITerminated
NCT00292045NY-ESO-1 protein + CpG 7909ICompleted[159]
NCT00140348GVAXI/IICompleted
NCT00140400GVAXI/IICompleted
NCT01095848DPX-0907ICompleted[160]
NCT00089856GVAXIIITerminated[161]
NCT00133224GVAXIIITerminated[162]
NCT00005039Fowlpox prostate specific antigenIITerminated
NCT00906243CV9103I/IITerminated[163]
NCT05104515OVM-200IRecruiting
NCT03384316ETBX-051 + ETBX-061 + ETBX-011ICompleted[164]
NCT03338790Nivolumab + Rucaparib
Nivolumab + Docetaxel + Prednisone
Novilumab + Enzalutamide
IIActive, not recruiting[156,157]
NCT03879122ADT + Docetaxel
ADT + Docetaxel + Nivolumab
ADT + Ipilimumab/Docetaxel + Nivolumab
II/IIIActive, not recruiting
NCT04382898 BNT112 +/− CemiplimabI/IIRecruiting[158]
NCT04077021CCW702ITerminated[159]
NCT03805594[177Lu]-PSMA-617 + PembrolizumabIbActive, not recruiting[160]
NCT04100018Nivolumab + Docetaxel + PrednisoneIIIRecruiting
NCT03637543NivolumabIIRecruiting[161]
NCT05580107MDPK67bIRecruiting
Table 6. Trials assessing exercise, diet, and microbiome in PCa patients.
Table 6. Trials assessing exercise, diet, and microbiome in PCa patients.
ClinicalTrials.gov IDInterventionPhaseStageReference
NCT03658486ExerciseN/A 1Recruiting
NCT03880422Aerobic and resistance exercise + dietN/ARecruiting
NCT02233608Advanced pelvic floor muscle exerciseI/IICompleted[168]
NCT01973673Bone health educational materialsN/ACompleted[169]
NCT05612880Physical function assessment following androgen receptor signalling inhibitorsN/ARecruiting
NCT03397030 *ExerciseN/ACompleted[170]
NCT00660686 *Resistance exercise + Flexibility trainingN/ACompleted[171]
NCT01696539SoC 2 + Walking interventionN/ACompleted[172]
NCT00658229Strength training groupIIICompleted[173]
NCT00253916 *Aerobic cardiovascular exercise
Resistance exercise
N/ACompleted[174]
NCT02453139Aerobic exerciseN/ACompleted[175]
NCT00329797Zoledronic acid and/or Calcium + Vitamin DIIICompleted
NCT02710721FastingN/ACompleted
NCT02946996Metformin + oligomeric procyanidin complexIIRecruiting
NCT03709485 *Correlation between microbiota and development of prostate cancerN/AUnknown[176]
NCT04687709 *Correlation between microbiome and ADT-related metabolic changesN/ARecruiting[177]
NCT04638049 *Correlation between microbiota/metabolome and radiation-induced gastrointestinal toxicitiesN/ACompleted
1 Not applicable; 2 standard of care; * not specified as advanced prostate cancer.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Puig-Saenz, C.; Pearson, J.R.D.; Thomas, J.E.; McArdle, S.E.B. A Holistic Approach to Hard-to-Treat Cancers: The Future of Immunotherapy for Glioblastoma, Triple Negative Breast Cancer, and Advanced Prostate Cancer. Biomedicines 2023, 11, 2100. https://doi.org/10.3390/biomedicines11082100

AMA Style

Puig-Saenz C, Pearson JRD, Thomas JE, McArdle SEB. A Holistic Approach to Hard-to-Treat Cancers: The Future of Immunotherapy for Glioblastoma, Triple Negative Breast Cancer, and Advanced Prostate Cancer. Biomedicines. 2023; 11(8):2100. https://doi.org/10.3390/biomedicines11082100

Chicago/Turabian Style

Puig-Saenz, Carles, Joshua R. D. Pearson, Jubini E. Thomas, and Stéphanie E. B. McArdle. 2023. "A Holistic Approach to Hard-to-Treat Cancers: The Future of Immunotherapy for Glioblastoma, Triple Negative Breast Cancer, and Advanced Prostate Cancer" Biomedicines 11, no. 8: 2100. https://doi.org/10.3390/biomedicines11082100

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