Next Article in Journal
MRI-Cavernosography: A New Diagnostic Tool for Erectile Dysfunction Due to Venous Leakage: A Diagnostic Chance
Previous Article in Journal
Preliminary Prognostication for Good Neurological Outcomes in the Early Stage of Post-Cardiac Arrest Care
Previous Article in Special Issue
Evaluation of Contrast-Enhanced Mammography and Development of Flowchart for BI-RADS Classification of Breast Lesions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Vaccines in Breast Cancer: Challenges and Breakthroughs

Division of Pharmaceutical Chemistry, Faculty of Pharmacy, Babu Banarasi Das Northern India Institute of Technology, Lucknow 226028, Uttar Pradesh, India
*
Author to whom correspondence should be addressed.
Diagnostics 2023, 13(13), 2175; https://doi.org/10.3390/diagnostics13132175
Submission received: 17 April 2023 / Revised: 9 June 2023 / Accepted: 21 June 2023 / Published: 26 June 2023

Abstract

:
Breast cancer is a problem for women’s health globally. Early detection techniques come in a variety of forms ranging from local to systemic and from non-invasive to invasive. The treatment of cancer has always been challenging despite the availability of a wide range of therapeutics. This is either due to the variable behaviour and heterogeneity of the proliferating cells and/or the individual’s response towards the treatment applied. However, advancements in cancer biology and scientific technology have changed the course of the cancer treatment approach. This current review briefly encompasses the diagnostics, the latest and most recent breakthrough strategies and challenges, and the limitations in fighting breast cancer, emphasising the development of breast cancer vaccines. It also includes the filed/granted patents referring to the same aspects.

1. Introduction

Cancer is a heterogeneous disease [1,2,3,4,5,6] with a poor median survival rate [3]. According to the WHO, it is the second-leading cause of death worldwide [5,6], with breast cancer (BC) being the most common form diagnosed in females [7]. About 5–10% of patients diagnosed with BC exhibit its metastatic form [8]. Moreover, it is highly challenging to forecast the prognosis of the illness with high certainty [3]. BC is classified as invasive or non-invasive [9,10]. The invasive form includes infiltrating ductal carcinoma (IDC) and invasive lobular carcinoma (ILC), while the non-invasive form includes ductal carcinoma in situ (DCIS) and lobular carcinoma in situ (LCIS) [10]. BC is further categorised depending upon the expressing hormone receptor such as the estrogen receptor (ER+), the human epidermal growth receptor 2 (HER2+), the progesterone receptor (PR+) and triple-negative breast cancer (TNBC), i.e., ER, PR, and HER2-all negative [11]. TNBC makes up 10–30% instances of BC [12] and is distinguished by a higher rate of relapse, higher potential for metastasis, and a shorter overall survival [13]. Additionally, cases of male BC, which may be either congenital, developmental, or acquired [14], have recently increased by about 40%, outpacing female cases by 25% of the affected population and by 18% in terms of mortality [15,16], though accounting for fewer than 1% of total BC diagnoses [14,17,18,19].
The article discusses at length the principle, approaches, and types of vaccines; and the routes of administration, antigens, and adjuvants used in the development of a BC vaccine, including combination therapeutic approaches and the clinical trials taking place in the field, along with a brief discussion on the diagnostics and the treatment strategies.

2. Diagnostics and Treatments in BC

In the recent past, various diagnostics have been developed for the detection of cancer in human subjects. The review briefly presents the diagnosis and its advancements, especially in BC detection. These include computer aided diagnosis (CAD), magnetic resonance imaging (MRI), tomography, Raman imaging, mammography, biopsy, radiomics, pathomics, and the use of artificial intelligence (AI), exosomes, and biomarkers.
CAD is a technique that is helpful in detecting cancer in tissue samples and distinguishes healthy from unhealthy tissue [20]. It also classifies cancer stages [21] using artificial intelligence. A digital multiple classifier database, called the Mammographic Image Analysis Society (MIAS), is a recent advancement for the better classification of lesions in mammograms [22,23]. MRI is a potential technique that is helpful in determining, with fineness and accuracy, the size and vascularisation of a tumour tissue [24]. It also provides the best soft tissue resolution [25] and, thus, has become a widely used tool for the in vivo characterisation of BC, which reduces the need for unnecessary biopsies [26]. Related to the tomographic technique, computed tomography (CT) in combination with positron emission tomography (PET) has become more crucial in the staging and restaging of BC [27]. The detection of distant metastases, using 18F-fluorodeoxyglucose positron-emission tomography (FDGePET/CT), was recently reported to have good sensitivity and specificity [28]. Along with MRI, a CT scan is more frequently utilised and offers a greater imaging resolution [29], with a high accuracy of up to 98% [27]. Raman spectroscopy is a quick and non-destructive optical technique [30] that does not require any staining process [31,32] for the diagnosis of human cancer [33]. The major advantage it offers is a label-free way of evaluating biological samples with great molecular specificity [34]. Despite the advanced diagnostics, conventional mammography still plays an important role in detecting BC. It is the art of capturing a picture of the breast [35] using a low-dose X-ray [20,36,37,38,39]. A combination of mammography with MRI or ultrasonography further improves the chances for the early detection of BC [40]. The technological advancement of mammography resulted in the following two forms: contrast-enhanced mammography [41] and contrast-enhanced spectral mammography. These techniques help to improve the sensitivity towards providing the detailed functional particulars of the anatomic and morphologic characters, as compared to a conventional mammogram [42,43]. When a mammogram or other imaging modality identifies some kind of abnormality, it is most likely that the technique called biopsy works. This includes surgical, core, and fine-needle aspiration [20]. Image-guided biopsy, through the utilisation of advanced interventional radiology techniques, improved the accuracy and safety of the procedure [44]. Another technique used is radiomics, which involves the extraction of information from the images obtained from high-dimensional imaging biomarkers. The technique helps to study the mechanisms occurring at the genetic and molecular levels for predictive and prognostic modeling [45,46]. It also enables the quantitative measurement of intra- and intertumoural heterogeneity, thereby enabling analysis of the entire tumour volume [47]. Similar to radiomics but based on a machine learning program, pathomics is useful for the study of breast oncology. It largely employs in vivo/ex vivo imaging, and analysis of the digital microscopy images of tissue, cells, and subcellular structures [46]. An added advantage is that it quantitatively assesses the structural features at multiple magnifications to complement the traditional histopathologic evaluation, with improved prediction of biological behaviour and clinical outcomes to guide treatment strategies [48].
In addition to the above diagnostic methodologies, the latest advancement in recent years is the use of machine learning programs. This has gained enormous popularity in the field of clinical BC research [3]. With its exceptional precision, AI is increasingly being suggested for use in BC screening [49], diagnosis, and prognosis [3,50]. The use of AI in BC risk assessment and prediction is also possible [16]. This uses digital mammography and digital breast tomosynthesis for detection [50,51,52]. Tomosynthesis involves removing the top layers of breast tissue for the ease of visualisation of the features and boundaries of the lesions [41]. AI is expected to be widely used in this field in the near future [52].
Exosomes are membrane-bound extracellular nanovesicles of endocytic origin [53,54,55]. They are present in diverse body parts [56,57] and may be extracted from extracellular fluids [55]. They play a crucial role in the progression of BC and are involved in the stimulation of tumour angiogenesis, the reorganisation of the stroma, and the promotion of tumour growth and create the tumour microenvironment (TME) [58]. Studies showed that exosome contents reassemble with cancer [54], thereby offering hope for lowering malignant cell activity and enhancing cancer imaging, prognosis [55], and liquid biopsy [53]. In addition, the expression pattern of exosomal microRNAs (miRNAs) correlates with the degree of tumour malignancy and prognosis. Therefore, circulating exosome-encapsulated miRNAs present an early prognostic biomarker for BC [58]. Any quantitative signs that reveal the presence or potential of malignancy or forecast tumour behaviour or prognosis are referred to as biomarkers [59]. Studies showed that the presence and the number of axillary node metastases are the most significant prognostic biomarkers in cases of BC. The ER marker is one of the BC prognostic biomarkers that is now available [60]. Currently, the multiple reaction monitoring (MRM) assay has been evolved to detect BC biomarker peptides in serum samples [61]. miRNAs and exosomes are new diagnostic and therapeutic biomarkers for BC patients [57,59]. Some biomarkers for BCB (breast cancer bone) metastases include CST1/2/4/6, PLAU, PLAT, COL6A1, and PLOD2. The advancement of technology, such as deep sequencing, lent advantages over conventional biomarkers by being non-invasive [59]. Proteins such as ER, ERR, Her2, Ki67, CEA, and TSGF and miRNAs such as miR-10b, miR-21, miR-145, and miR-155 are examples of distinct BC biomarkers [57].
The treatment of cancer has always been a challenge for the scientific community. The following paragraphs briefly describe some of the strategies developed over time to treat BC.
One of the most advanced strategies for the treatment of BC involves the use of nanoparticles (NPs). They are an efficient means to deliver medications targeting malignant cells [62], with improved pharmacokinetics and pharmacodynamics [63] and reduced side effects [64]. For example, a liposomal nanoformulation of Doxorubicin was shown to display action with significantly reduced side effects. Catechols, gallol, and their derivatives are used to construct nanomaterials [65], with the use of polymers, to deliver conjugated or encapsulated drugs [66]. Gold, silver, and iron oxide NPs are thermally stable, low in toxicity, compact, and effective in BC treatment [11]. In addition, magnetic NPs are also being studied as promising materials for prospective nanopharmaceuticals [67]. Neoadjuvant treatment is an effective treatment strategy for inoperable locally advanced BC [68]. In patients with non-metastatic BC and axilla tumours, neoadjuvant therapy is used to de-escalate the extent of surgery [69], thereby minimising unnecessary surgeries and mastectomies [70]. In order to fix, add, or repress a gene, the genetic material must be first administered through a vector to the target cells. Gene therapy is less toxic than conventional medicine and helps to target tumour cells without harming normal cells [71].
Radiotherapy (RT) exerts local action [72] and represents one of the most effective non-surgical treatment modalities [73]. Whole breast irradiation, also known as WBI, is frequently carried out following a breast-conserving surgery [29], i.e., intraoperative radiation therapy (IORT), which is a therapeutic strategy that accomplishes surgery and adjuvant RT in a single treatment [29].
The dynamic relationship between tumour cells and the immune system involves three key stages: elimination, equilibrium, and escape [74]. Immunotherapy (IT) is a strategy that works by stimulating and recruiting a patient’s immune system against the tumour cells [75,76,77]. Various IT approaches include tumour-targeting antibodies, adoptive T-cell therapy, vaccines, and immune checkpoints [60]. Glycoprotein mucin-1 (MUC-1) was one of the first BC-associated antigens to be identified [78]. In order to evaluate the cell–cell interactions, the use of bioengineering tools such as optical microscopy and a microphysiological in vitro system was accomplished [79]. IT, however, can negatively affect the foetus or impede a future desired pregnancy [80].
The use of combination therapy may give a new hope for BC treatment [81,82]. It is a promising therapeutic approach that involves folate ligands in conjunction with chemotherapeutic drugs and small interfering RNAs (siRNAs) [83]. A study undertaken by Ma et al. reported the beneficial effects of a combination of Trastuzumab and Everolimus in patients with ER+/HER2-HER mutant BC [84]. It reported a boost in the survival rates of patients by up to 35%. Some examples of combination approaches include the combinations of any of the following therapies: immunotherapy, molecular-targeted therapy, cell cycle management, cell signalling pathway regulation, monoclonal antibodies, and antibody–drug conjugates [60].
Thermal ablation (TA) therapy exerts its action by destroying the entire tumour using heat, through needle-like applicators [85]. It results in minimal invasion without damaging the adjacent vital structures [86,87] and, thus, is a safe and effective alternative to surgical resection in early-stage treatment of BC [88]. This advancement resulted in percutaneous TA techniques, viz., radiofrequency ablation, microwave ablation, cryotherapy [89], and cryoablation [90,91]. Another method is radiofrequency ablation (RFA), which entails inserting a metal electrode into the breast and connecting it to a radiofrequency generator that outputs 200 W at 400–800 KHz. [91]. The tumour tissue is burned at temperatures (50 °C) high enough to cause necrosis [92]. Microwave (MW) ablation is a recent advancement in the field that involves heating malignant tissue with MW energy to cause cell death [91]. The goal of hyperthermic therapy is to kill tumour cells by inducing a series of metabolic changes, such as apoptosis in the tumour tissue by heating it to high temperatures [93]. The use of magnetic nanoparticles (MNPs) such as iron oxide nanoparticles (IONPs), in an alternating magnetic field results in magnetic hyperthermia. The technique utilises two types of magnetic nanoparticles: magnetic alloy nanoparticles (MANPs) and magnetic metal oxide nanoparticles (MMONPs) [94]. They are site-specific and are classified as whole-body hyperthermia, regional hyperthermia, and localised hyperthermia depending on the site of application [95].
In addition, modern medicine emphasises the prevention and treatment of cancer with natural nutritional components [96]. This includes the use of flavonoids, phenolic compounds, and some dietary sources that effectively exhibit anticancer activity [96,97,98,99]. For example, quercetin prevents free radicals from impairing low-density lipoproteins and is useful against cancer [96]. Luteolin [99] and resveratrol proved to modulate various signaling pathways involved in the proliferation of cancerous cells and are established as anticancer agents [100]. Some naturally occurring constituents such as the ϖ-3 fatty acids and phytoalexin found in grapes; epigallocatechin gallate, a polyphenolic compound found in herbs such as Oldenlandia diffusa and Ziziphus jujube [98]; and Silibinin, a major component of Silymarin that is obtained from the seeds of milk thistle, Silybum marianum [97], can be used in the treatment of BC. Pomegranate, Punica granatum, can also be used against different type of cancers [101].

3. Vaccine Therapy in BC

3.1. Introduction

Vaccines have long protected humans from communicable and non-communicable diseases [102]. Therefore, conventionally, the word “vaccine” is usually related to the fight against infectious diseases. Vaccines exert their action by stimulating immune responses. This is achieved by inoculating a healthy individual with attenuated/detoxified bacteria, viruses, or extracted toxins [103].
The immune system works to keep living things in a state of equilibrium by the process called immune surveillance [104]. The method by which tumour cells circumvent the immune system has been extensively explored and has been successfully established over the past few years. Such efforts led to the conclusion that cancer immunoediting is the strategy employed by the tumour cells towards immune evasion [105,106]. It may be caused by TME-antigen-mediated antitumour immunological responses. A number of tools for cancer immunotherapy were developed, which include antibodies, peptides, proteins, nucleic acids, and immune-competent cells such as dendritic cells and T-cells [103]. Cancer immunotherapies are now considered the fourth treatment method [107,108], used either alone or in combination [60]. The therapy was initially utilised by William B. Coley in 1891, to treat sarcoma patients with Coley’s toxin [109].
The available vaccines for cancer immunotherapy can be divided into two basic types, the prophylactic and the therapeutic vaccines [110,111]. The former induces immunological memory by vaccinating healthy people [112], to prevent morbidity from a certain malignancy [113], and can be a cost-effective preventive measure [114]. The latter boosts immune systems in people detected with cancer [103]. The therapeutic cancer vaccine prevents the growth of advanced malignancies or relapsed tumours that are resistant to standard treatments [112]. Based on the structure and the content, the vaccines are further classified into the cell vaccines, the peptide vaccines and the nucleic acid vaccines [103].
The criteria necessary to be fulfilled to achieve vaccination requires a target antigen on tumour cells to stimulate the immune response, a vector to deliver the vaccine-derived antigen to the immune system, an adjuvant to boost immunological stimulation, and an appropriate monitoring tool [115]. These are discussed in detail in the subsequent sections.

3.2. Concepts in Designing of a Breast Cancer Vaccine (BCV)

3.2.1. Immunoediting

The immune system constantly changes as BC progresses. The process is called immunoediting, which comprises three steps: elimination, equilibrium, and escape. During elimination, the tumour cells stimulate the innate immune system (which along with the adaptive immune response can recognise and remove early altered tumour cells) through the activation of macrophages, natural killer cells (NK-cells), and dendritic cells (DCs), in turn activating the tumour-targeted T-lymphocytes. The equilibrium phase initiates in the case of a cancer subclone colony surviving the host’s immunity. This stage creates a delicate balance between cancer growth and the immune system’s defence function, making it difficult to totally remove the tumour cells, though their progression is severely limited [116]. However, it results in the formation of cancer cells with decreased immunogenicity, epigenetic alterations, and genetic instability [117], thus making them capable of escaping the immune detection and destruction [118]. Such cells that escape the immunological pressure finally enter the third stage of immunoediting, where the immune system barely puts any restraints on the progression of the modified tumour cells as in Figure 1 [119].

3.2.2. Immune Surveillance

Effector immune cells must directly recognise tumour antigens from tumour cells or indirectly from antigen-presenting cells (APCs), via the major histocompatibility complex (MHC) on the cell surface, to initiate an immune response. CD8+ and CD4+ T-cells are critical to the immunoediting process and help separate the non-self epitopes of the tumour cells expressed by MHC class I and MHC class II molecules from the normal self-antigens [120,121]. Tumour-specific antigens (TSAs) and tumour-associated antigens (TAAs) [122] are examples of tumour antigens. These include germ-line antigens, tissue differentiation antigens, and overexpressed antigens, exemplified by the melanoma-associated antigen, the carcino-embryonic antigen (CEA), and HER2 and MUC-1, respectively [123]. Many of the tumour antigens employed in immunotherapy are expressed in normal tissues as well. However, tumour cells overexpress these antigens [122].

3.2.3. Immune Suppression

Tumour cells successfully inhibit the host’s immune system, both locally and systemically, from evading immune surveillance [124]. It is well-established that there is predominance of the immunosuppressive effect with advancement of the disease. This results in a gradual transition from the elimination phase to the escape phase [125]. During this process, there is an alteration of the number of cells around TME and the lymph nodes in the vicinity of the tumour tissue. First are regulatory T-cells (Treg cells). There is enhancement in the proliferation of Treg cells brought about by transforming growth factor-β (TGF-β) [126], while cytotoxic T-lymphocytes (CTLs) are greatly reduced, as interleukin-2 preferably binds to Treg cells. The antitumour response is, thus, weakened by a reduction in the number of CTLs [127] and NK-cells [128] in TME (Figure 2). Treg cells function to downregulate the dendritic cell co-stimulatory markers, viz., CD80 and CD86, necessary for the priming of CTLs [129], which, in turn, function to eliminate any abnormal-phenotype-expressing cell. Second are tumour-associated macrophages (TAMs). TAMs release inhibitory cytokines including IL-10 and TGF-β to suppress CTL activity and the production of IL-12 [130]. Third are the myeloid-derived suppressor cells (MDSCs). During the shift, these cells start appearing in the peripheral blood as well [131]. Tumour cells also activate immunological checkpoint receptors such as cytotoxic T-lymphocyte antigen-4 (CTLA-4) and programmed cell death receptor-1 (PCDR-1) [132]. PCDR-1 blocks programmed cell death ligand-1 (PCDL-1).
Therefore, in patients with diverse malignant tumours, the tumour-infiltrating lymphocytes (TILs) and the tumour-specific T-lymphocytes show high amounts of PCDR-1. The involvement of PCDL-1/PCDR-1 by tumour cells prevents the elimination of T-cells, thereby causing their dysfunction and, ultimately, cell death. In contrast, the cytokines that create an immunosuppressive milieu help to further promote tumour growth [133].

3.2.4. Identification of the Antigen for BC Immunotherapy

Numerous tumour antigens, which are expressed in healthy cells but are overexpressed in tumour cells, are employed in BC immunotherapy. They include HER2, p53 (tumour protein 53), MUC1, carcinoembryonic antigen (CEA), telomerase reverse transcriptase (hTERT), and carbohydrate antigens [78]. Due to their widespread expression in the majority of tumour types, some of these antigens are known as universal tumour antigens. An example of this class is hTERT [134]. All the potential antigens that are used in the creation of vaccines for the management of BC are briefly discussed in the following section.

Human Epidermal Growth (HER2) Receptor 2

HER2 is a tyrosine kinase that regulates cell proliferation and survival. The oncogene for HER2 is located on chromosome number 17q12. In cases where BC is HER2-positive, an amplification of 15–20% in the expression of HER2 is found. This is responsible for triggering the growth and progression of tumour cells [135]. HER2 is linked to a more severe form, a higher risk of recurrence, and a higher mortality rate. The first drug to be established as a HER2 blocker was Trastuzumab [136], which has been validated for the treatment of patients with HER2+ BC [137,138].

Tumour Suppressor p53 Protein (p53)

The gene encoding p53 is located on chromosome number 17 [139]. It plays a vital role in the maintenance of DNA integrity and the prevention of cancer. Under normal physiological conditions, when a cell’s DNA is damaged, there is an induction of the p53 protein that causes arrest of the cell cycle. This allows cells to repair themselves, but, if the damage is too severe, the cells apoptise and are rejected. In a variety of cancer forms, mutations in the p53 gene (mtp53) takes place [140]. However, mtp53 is less frequently found in BC [141], though it has high significance in the diagnosis and prognosis of TNBC, with almost 70%–80% of cases displaying mtp53 [142].

Mucin1 (MUC1)

The MUC1 gene is encoded by chromosome 1q21. It is a high molecular weight transmembrane glycoprotein that functions as a physical barrier to protect the epithelial layer of cells from environmental exposure. It forms the epithelial lining of the respiratory and GI tracts, mammary glands, pancreas, liver, and kidneys. MUC1 is a poly-morphic type I member of the mucin family. It is overexpressed in about 90% of human BC due to genetic alterations and the dysregulation of transcription [143,144]. MUC1 is also aberrantly glycosylated [145], which exposes various antigens, thereby generating a new set of antibodies that can prove beneficial in the diagnosis of cancer [146].

Carcinoembryonic Antigen (CEA)

The CEA is a glycoprotein that is encoded by human chromosome 19q13.2. This antigen is an important serum biomarker for the detection of cancer and plays a vital role in the prognosis and diagnosis of BC. It is found elevated in patients with metastatic BC, especially with bone metastasis [147,148]. Several studies suggested the significance of CEA and CA15-3 levels in predicting BC patients who can be operated on at an early stage [149].

Human Telomerase Reverse Transcriptase (h-TERT)

The gene encoding h-TERT, a catalytic subunit of the telomerase, is encoded on chromosome 5p15.33. This ribonucleoproteic enzyme is responsible for synthesising telomeres, which, in turn, maintain the chromosomal length. The ultimate result is cellular immortalisation [150]. Studies suggested their vital role in the development of cancer. It was found that in most forms of cancer, the reactivation of telomerase takes place in a dependent or an independent manner. The enzyme ensures the stability of chromosomes, thus bypassing senescence. TERTp is the promoter region of TERT. Mutations in the TERT gene support carcinogenesis with variable frequencies, which takes place through the healing of the telomere’s length, thus expanding the life of cells. TERTp was established as a tool to characterizes the type of cancer [151] and, thus, is helpful in the diagnosis and prognosis of the disease [152].

3.3. Design Approaches of a BCV

The optimisation of vaccine schedules and administration methods is a component of a vaccination strategy. Based upon the platforms and formulations, BCVs can be broadly classified as peptide- and protein-based [153], carbohydrate antigen-based, whole-cell-based, gene-based [154], and fusion-based vaccines [155]. However, irrespective of the type, all vaccines are dependent on the autologous immune system recognising a specific antigen to exert a therapeutic effect. Moreover, the use of an adjuvant is essential, as it helps to increase the antigen immunogenicity, thereby controlling the immune response [156].

3.3.1. Peptide- and Protein-Based Vaccine (PV)

In this class of BCVs, the MHC class I restricted peptide epitopes are used to stimulate the immune response against a tumour antigen as shown in Figure 3 [157]. The injected peptide stimulates immune effector cells to find and kill cancer cells [158]. Some short amino acid peptides are preferably used, as they are cheap, stable, and easy to synthesise and modify and display low immunogenicity [159]. However, a vaccination cannot be given to patients with a non-common human leukocyte antigen type, since each peptide is confined to a specific HLA subtype.
MHC class I binding peptides poorly stimulate CD4+ helper T-cells and also, in turn, limit their ability to activate CD8+ cytotoxic T-cells, resulting in a transitory immunological response. According to Pallerla et al., long peptides are capable of containing many MHC class I and class II epitopes, thus helping to partially overcome this difficulty. Such peptides containing 23–45 amino acids may improve T-cell activation through processing and presentation [154]. The success of the peptide-based BCV can be seen from its entry into phase I/II of clinical trials [160].
The entire tumour antigen protein or a truncated portion of it, wherein the sequence of amino acids is substantially longer than that of peptides, is used to create protein-based vaccines [161]. It is not HLA-restricted and allows for the absorption, processing, and presentation of a variety of MHC class I and class II peptide epitopes [162]. However, the presenting method may be less effective, and the lack of a precise marker makes it difficult to estimate how well such vaccine types would work [163]. A number of clinical trials (Table 1) are currently in the pipeline to test the efficacy of vaccines pertaining to BC [162].

3.3.2. Carbohydrate Antigen-Based Vaccine (CAV)

Immune cells can discriminate between improperly expressed carbohydrate antigens in tumour cells. This makes the carbohydrate antigen a prime candidate for inclusion in cancer vaccination. An example includes the expression of a unique disaccharide carbohydrate, Sialy-Tn (STn), on the cell surface of cancer cells, including BC cells, which is connected to MUC-1 [164]. According to Munkley (2016), immunisation with STn results in tumour regression and prolongation of survival time. Thus, it could be beneficial in the development of a cancer vaccine [165].

3.3.3. Whole Tumour Cell-Based Vaccine (WTCV)

One of the conventional approaches in the development of a cancer vaccine is the stimulation of the immune response by the use of whole tumour cells or the products obtained from tumour cell lysis. A WTCV induces a polyvalent immune response, since it is based on a pool of unknown antigens created by autologous or allogeneic tumour cells [166]. Sometimes, the enhancement of the antigen-presenting ability of the WTCV is achieved through engineered tumour cells that are capable of releasing cytokines or expressing co-stimulatory molecules. The drawback of the WTCV is that it contains endogenous cellular antigens, which can result in an autoimmune response. A systematic procedure for creating the WTCV is, however, lacking [167].

3.3.4. Dendritic Cell-Based Vaccine (DCV)

Upon migration into the lymph nodes, a diverse population of APCs, called DCs, effectively absorb antigens to process and present them to CD4+ and CD8+ T-cells. DCs can also stimulate NK-cells and B-cells. DC-based vaccines typically use ex vivo generated DCs that were transfected to express tumour antigens or are loaded with tumour antigens. In a review by Butterfield et al., the use of antigens, such as complex tumour lysates and several MHC class I and class II peptides, and monocytes and CD34+ progenitor cells for the purpose of DCV preparation was discussed [57]. The technical challenge that arises while developing DC-based vaccines was because of the distinct process of ex vivo DC maturation [168].

3.3.5. Gene-Based Vaccine

The strategy for a gene-based vaccine involves a plasmid that carries the DNA encoding the cancer antigen. This form of vaccine can be used to activate both a non-specific innate immunity and an adoptive immunity specific to an antigen. Due to its simplicity, safety, and cost-effectiveness, this approach is regarded as one of the most practicable methods for cancer immunotherapy. However, it suffers from limitations such as a lack of plasmid uptake and ineffective antigen expression, thus presenting insufficient immunogenicity [162]. To overcome this demerit, various approaches were undertaken. One of the approaches was to make the vaccine self-replicating. This utilised the RNA replicase encoding gene [169]. RNA-based medications have the potential to be effective pharmacological regulators against cancer cells by altering the expression of particular proteins. These characteristics help to increase specificity and reduce the chance of off-target impacts [153].

3.3.6. Fusion Vaccine

This strategy employs the fusion of autologous DCs and autologous whole tumour cells. It involves the cytoplasm of both cell types to be fused together without their nuclei doing so. This preserves their ability to function as individual cells. With the fusion, the formed vaccine can express and process a wide range of recognised and unrecognised tumour antigens [170]. Studies and trials found a favourable response by patients with metastatic BC towards this fusion vaccine, in terms of its potent antitumour effects [154,171].

3.4. Adjuvants Used in Design of BCVS

Adjuvants are combined with the antigen and are essential in the case of low immunogenicity. They help to increase the immunogenicity of the antigen and thus, trigger, the immune response, particularly in the elderly [156]. Most adjuvants work by decreasing antigen release, encouraging antigen absorption and presentation by APCs, and boosting the growth of DCs and macrophages [172]. Traditional adjuvants, such as alum, mostly stimulate type-2 T-helper cell-dependent humoral immunity in prophylactic vaccinations for infectious diseases rather than type-1 T-helper cell responses that directly destroy tumour cells [173]. A common adjuvant in BCV is a secreted cytokine, called granulocyte-macrophage colony-stimulating factor (GM-CSF). It was demonstrated to promote the proliferation and activation of DCs as well as the maturation of myeloid cells such as granulocytes and macrophages [174,175]. Clinical trials of many GM-CSF-containing BCVs revealed measurable immune responses. The local administration of GM-CSF to melanoma patients increases the probability of the antigen immune response after vaccination. Studies found GM-CSF suppresses T-cell responses and produces inhibitory MDSCs. However, elaborative research is needed to discover the role of GM-CSF as an adjuvant for cancer vaccines.
DNA-based cancer vaccines also use recombinant viral vector adjuvants (Figure 4). Recombinant viral vectors, which commonly carry antigens, contain different levels of pattern recognition receptor (PRR) and toll-like receptor (TLR) ligands that activate DCs and boost immune response. TLR agonists activate CD8+ T-cells and prevent T-cell exhaustion. The vectors contain other sequences that can compete with the targeted antigen motif, which is the main drawback of this adjuvant. However, adjuvant effects vary depending on vaccination formulation, targeted tumour antigens, immunisation schedule, and mode of administration, making adjuvant comparisons difficult [176]. Moreover, adjuvant optimisation tests for BCV are crucial.

3.5. Routes of BCV Administration

A good vaccine for cancer should be capable of efficiently transferring antigens to autologous APCs. For this purpose, various strategies are preferred. HER2 peptide-based vaccines are usually intradermally administered and exhibit an enhanced rate of response. This is probably due to the widespread network of DCs. Low intradermal peptide doses are safe and trigger specific responses by antigen T-cells in most healthy human subjects [177,178,179]. Several BCVs were tested for their immune response induction efficiency via subcutaneous injection [180]. However, a large dose of antigen at the site of injection may cause severe reaction and sporadic sterile abscesses, which may require vaccine discontinuation or a dosage reduction [175]. Vectors or plasmids are injected through various routes in the case of the administration of a DNA-based vaccine, of which the intramuscular route of administration was found to display the most effective immune response [181]. Some DC-based immunisations must be given intravenously to directly stimulate lymph node T-cells [157].

3.6. Clinical Trials of BCVs

In preliminary studies, certain BCVs were successful in eliciting discernible immune responses and showing good tolerance. However, most of them showed appreciable clinical advantages in the ensuing Phase 3 trials. Recently, the NeuVaxTM vaccine (developed by Galena Biopharma, a US-based biotechnology company), which is administered with Leukine® for the disease condition of BC with intermediate to low HER2 expression, was granted a Special Protocol Assessment for its Phase 3 trial (Prevention of Recurrence in Early-Stage, Node-Positive Breast Cancer) [182]. The Theratope® and Enhanzyn™ vaccines are some of the vaccines that were withdrawn from the clinical trial enrolment [183]. A brief summary of the vaccines in clinical trials is listed in Table 1.

3.7. Combinational Therapy of BCVs

Immune checkpoint blockers (ICB) have changed the approach towards the treatment of cancer. As far as BC is concerned, ICBs were already proven to be effective in treating metastatic TNBC [184]. The addition of ICB to Trastuzumab, however, was linked to additional side events and did not provide a clinically substantial improvement in the progression-free survival for HER2-positive metastatic BC [185]. Combining the vaccination with ICB to combat cancer tolerance is a trending research approach in the field [186]. As previously stated, ICB blocks inhibitory receptors such as PCDR-1/PCDL-1 and CTLA-4 to enable the effector immune cells to kill tumour cells. According to certain preclinical research, when T-cells are activated by tumour vaccines, the inhibitory receptor expression on the cell surface also increases. One of the underlying reasons is the enhanced interferon-γ (IFN-γ), which is released by tumour-specific T-cells. IFN-γ up regulates the expression of PCDL-1 on the tumour cells and APCs. PCDL-1 initially helps to prevent the body’s immune responses from being too amplified [187,188]. Therefore, the immunosuppressive impact that reduces the antitumour immunity elicited by vaccines is likely to be relieved by an injection of ICB [189]. A promising approach that has the potential to improve and lengthen the course of the immune response and effectively produce considerable clinical benefits is the combination of the BC vaccination with ICB. Additionally, combining cancer vaccinations with recognised medicines could also increase efficacy.
The research suggests that some HER2-derived peptide vaccines [190] and anti-HER2 monoclonal antibodies may function synergistically [191]. Studies show a connection between chemotherapy/radiation therapy and immune-related cell death. When these medicines are applied in conjunction with cancer vaccines, it might create a long-lasting immune response. Consistently, it would be worth investigating the effects of integrating cancer vaccination with chemotherapy [192,193], hormone therapy [194,195], targeted therapy [196,197], and radiation therapy [198,199].

3.8. Challenges Faced during the Course of Development of BCVs

Despite the several advantages associated with cancer vaccines, there are numerous challenges accompanying cancer vaccination strategies. Firstly, the development of tolerance towards the antigen, which results in lowered immune response. Secondly, the heterogenicity of the tumour type brings in some diverse intrinsic and extrinsic pressures, which, in turn, result in a detrimental effect on the antitumour vaccination processes. In addition, some genetic and non-genetic mechanisms play a role in defining the heterogenicity of cancerous cells, thus affecting the immune response. Moreover, the immune invasion mechanism was found to be a major hurdle in the efficacy of a vaccine. This requires potential alternative strategies to improve antitumour immunity [200]. Some developed vaccines were found to display poor immunogenicity when used alone. Therefore, the next generation of adjuvants may be used [201]. A key point during the development of an anticancer vaccine is the aftermath, in the form of immunological responses. Thus, it is necessary to recognise the immune modulatory pathway, and test and validate the disease-specific cancer vaccination [202]. Cancer immune interaction is also governed by the concomitant use of a drug.

4. Recent Patents Filed/Granted on Cancer Treatment and Diagnostics

This review also encompasses the patents filed/granted at the USPTO during 2018–2022, as shown in Table 2 [203,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218,219,220,221,222,223,224,225,226,227,228,229,230,231,232,233,234,235,236,237,238,239,240]. The patents present advancements in diagnostic technologies as well as in cancer therapeutics.

5. Conclusions

Breast cancer (BC) is one of the most prevalent diseases worldwide. However, advancements in science and technology have aided the development of methods for BC diagnosis. They have also transformed the way BC is treated. As far as BC therapeutics are concerned, a revolution was introduced by the use of the immune system as an instrument. This led to the development of several promising BCVs, including preventive, therapeutic, and fusion vaccines. Even though many strategies have been employed to date, developing a cancer vaccine still remains challenging. The major challenges faced when creating a cancer vaccine include inadequate immunogenicity, tumour heterogeneity, poor antigen identification, and the health consequences.
An overview of the approaches for diagnosing BC, related treatments, vaccines, and patents was included in the article to provide leads for future research. Thus, with the advancements in research tools and new technologies, cancer vaccines may soon be a reality.

Author Contributions

Conceptualization, S.K.S. and G.N.F.; Software, G.N.F. and H.F.; Resources, S.K.S., G.N.F. and H.F.; Writing-original draft preparation, G.N.F. and H.F.; Writing, review and editing, S.K.S., G.N.F. and H.F.; Supervision, S.K.S. All authors have read and agreed to the published version of the manuscript.

Funding

No funding was received for preparation of this manuscript.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

List of Abbreviations

AIartificial intelligence
APCsantigen-presenting cells
BCbreast cancer
BCVbreast cancer vaccine
CADcomputer aided diagnosis
CAVcarbohydrate antigen-based vaccine
CEAcarcino-embryonic antigen
CTcomputed tomography
CTLA-4cytotoxic T-lymphocyte antigen-4
CTLscytotoxic lymphocytes
DCISductal carcinoma in situ
DCsdendritic cells
DCVdendritic cell-based vaccine
ERestrogen receptor
FDGePET/CT18F-fluorodeoxyglucose positron-emission tomography
FZD receptorshuman Frizzled
GM-CSFgranulocyte-macrophage colony-stimulating factor
HER2human epidermal growth receptor2
h-TERTtelomerase reverse transcriptase
ICBimmune checkpoint blockers
IDCinfiltrating ductal carcinoma
IFN-γinterferon-γ
ILCinvasive lobular carcinoma
IORTintraoperative radiation therapy
ITimmunotherapy
LCISlobular carcinoma in situ
MANPsmagnetic alloy nanoparticles
MDSCsmyeloid-derived suppressor cells
MHChistocompatibility complex
MIASMammographic Image Analysis Society
miRNAsexosomal microRNAs
MMONPsmagnetic metal oxide nanoparticles
MNPsmagnetic nanoparticles
MRImagnetic resonance imaging
MRMmultiple reaction monitoring
mTOR kinasemammalian target of rapamycin kinase
mtp53mutation in the p53 gene
MUC-1mucin-1
MWmicrowave
NK-cellsnatural killer cells
NPsnanoparticles
p53tumour protein 53
PALPplacental alkaline phosphatase
PCDprogrammed cell death
PCDL-1programmed cell death ligand-1
PCDR-1programmed cell death receptor-1
PETpositron emission tomography
PRprogesterone receptor
PRRpattern recognition receptors
PVpeptide and protein-based vaccine
RFAradiofrequency ablation
ROR1-antibodiesreceptor tyrosine kinase-like orphan receptor1 antibodies
siRNAssmall interfering RNAs
STnSialy-Tn
TAthermal ablation
TAMstumour-associated macrophages
TCRT-cell receptor
TGF-βtransforming growth factor-β
TILstumour-infiltration lymphocytes
TLRtoll-like receptors
TMEtumour microenvironment
TNBCtriple-negative breast cancer
Treg cellsregulatory T-cells
TSA tumour-specific antigens
WTCVwhole tumour cell-based vaccine

References

  1. Dittmer, J. Breast Cancer Stem Cells: Features, Key Drivers and Treatment Options. Semin. Cancer Biol. 2018, 53, 59–74. [Google Scholar] [CrossRef] [PubMed]
  2. Fisusi, F.A.; Akala, E.O. Drug Combinations in Breast Cancer Therapy. Pharm. Nanotechnol. 2019, 7, 3–23. [Google Scholar] [CrossRef] [PubMed]
  3. Huang, S.; Yang, J.; Fong, S.; Zhao, Q.J. Artificial Intelligence in Cancer Diagnosis and Prognosis: Opportunities and Challenges. Cancer Lett. 2020, 471, 61–71. [Google Scholar] [CrossRef] [PubMed]
  4. Hylton, N.M.; Gatsonis, C.A.; Rosen, M.A.; Lehman, C.D.; Newitt, D.C.; Partridge, S.C.; Bernreuter, W.K.; Pisano, E.D.; Morris, E.A.; Weatherall, P.T.; et al. Neoadjuvant Chemotherapy for Breast Cancer: Functional Tumor Volume by Mr Imaging Predicts Recurrence-Free Survival-Results from the Acrin 6657/Calgb 150007 I-Spy 1 Trial. Radiology 2016, 279, 44–55. [Google Scholar] [CrossRef] [Green Version]
  5. Da Costa, B.R.B.; De Martinis, B.S. Analysis of Urinary Vocs Using Mass Spectrometric Methods to Diagnose Cancer: A Review. Clin. Mass Spectrom. 2020, 18, 27–37. [Google Scholar] [CrossRef]
  6. Hasan, M.; Büyüktahtakın, İ.E.; Elamin, E. A Multi-Criteria Ranking Algorithm (Mcra) for Determining Breast Cancer Therapy. Omega 2019, 82, 83–101. [Google Scholar] [CrossRef]
  7. Han, H.J.; Ekweremadu, C.; Patel, N. Advanced Drug Delivery System with Nanomaterials for Personalised Medicine to Treat Breast Cancer. J. Drug Deliv. Sci. Technol. 2019, 52, 1051–1060. [Google Scholar] [CrossRef]
  8. Claessens, A.K.M.; Ibragimova, K.I.E.; Geurts, S.M.E.; Bos, M.E.M.M.; Erdkamp, F.L.G.; Tjan-Heijnen, V.C.G. The Role of Chemotherapy in Treatment of Advanced Breast Cancer: An Overview for Clinical Practice. Crit. Rev. Oncol. Hematol. 2020, 153, 102988. [Google Scholar] [CrossRef]
  9. De Cicco, P.; Catani, M.V.; Gasperi, V.; Sibilano, M.; Quaglietta, M.; Savini, I. Nutrition and Breast Cancer: A Literature Review on Prevention, Treatment and Recurrence. Nutrients 2019, 11, 1514. [Google Scholar] [CrossRef] [Green Version]
  10. Subramani, R.; Lakshmanaswamy, R. Chapter Three—Pregnancy and Breast Cancer. In Progress in Molecular Biology and Translational Science; Elsevier: Amsterdam, The Netherlands, 2017; Volume 151, pp. 81–111. [Google Scholar] [CrossRef]
  11. Barzaman, K.; Karami, J.; Zarei, Z.; Hosseinzadeh, A.; Kazemi, M.H.; Moradi-Kalbolandi, S.; Safari, E.; Farahmand, L. Breast Cancer: Biology, Biomarkers, and Treatments. Int. Immunopharmacol. 2020, 84, 106535. [Google Scholar] [CrossRef]
  12. Bai, X.; Ni, J.; Beretov, J.; Graham, P.; Li, Y. Immunotherapy for Triple-Negative Breast Cancer: A Molecular Insight Into the Microenvironment, Treatment, and Resistance. J. Natl. Cancer Center 2021, 1, 75–87. [Google Scholar] [CrossRef]
  13. Huang, M.; Zhang, J.; Yan, C.; Li, X.; Zhang, J.; Ling, R. Small Molecule Hdac Inhibitors: Promising Agents for Breast Cancer Treatment. Bioorg. Chem. 2019, 91, 103184. [Google Scholar] [CrossRef] [PubMed]
  14. Swamy, N.; Rohilla, M.; Raichandani, S.; Bryant-Smith, G.J. Epidemiology of Male Breast Diseases: A 10-Year Institutional Review. Clin. Imaging 2021, 72, 142–150. [Google Scholar] [CrossRef]
  15. Konduri, S.; Singh, M.; Bobustuc, G.; Rovin, R.; Kassam, A. Epidemiology of Male Breast Cancer. Breast 2020, 54, 8–14. [Google Scholar] [CrossRef]
  16. Sheth, D.; Giger, M.L. Artificial Intelligence in the Interpretation of Breast Cancer on Mri. J. Magn. Reson. Imaging 2020, 51, 1310–1324. [Google Scholar] [CrossRef] [PubMed]
  17. Chesebro, A.L.; Rives, A.F.; Shaffer, K. Male Breast Disease: What the Radiologist Needs to Know. Curr. Probl. Diagn. Radiol. 2019, 48, 482–493. [Google Scholar] [CrossRef]
  18. Shaaban, A.M. Pathology of the Male Breast. Diagn. Histopathol. 2019, 25, 138–142. [Google Scholar] [CrossRef]
  19. Shin, K.; Whitman, G.J. Clinical Indications for Mammography in Men and Correlation with Breast Cancer. Curr. Probl. Diagn. Radiol. 2021, 50, 792–798. [Google Scholar] [CrossRef]
  20. Jalalian, A.; Mashohor, S.; Mahmud, R.; Karasfi, B.; Saripan, M.I.B.; Ramli, A.R.B. Foundation and Methodologies in Computer-Aided Diagnosis Systems for Breast Cancer Detection. EXCLI J. 2017, 16, 113–137. [Google Scholar] [CrossRef]
  21. Liew, X.Y.; Hameed, N.; Clos, J.J.C. A Review of Computer-Aided Expert Systems for Breast Cancer Diagnosis. Cancers 2021, 13, 2764. [Google Scholar] [CrossRef]
  22. Alkhateeb, A.J.J. Breast Cancer Computer-Aided Diagnosis System from Digital Mammograms. J. Adv. Med. 2019, 30, 530197. [Google Scholar] [CrossRef] [Green Version]
  23. Al-Shamlan, H.; El-Zaart, A. Breast Cancer Computer Aided Diagnosis (Cad) System. In Image Processing, Computer Vision, and Pattern Recognition; Kindle Direct Publishing: Seattle, WA, USA, 2011; p. 1. [Google Scholar]
  24. Chirita, A.J. Indications of the Magnetic Resonance Method in Breast Pathology. Chirurgia 2017, 112, 367–377. [Google Scholar] [CrossRef] [PubMed]
  25. Guneyli, S.; Erdem, C.Z.; Erdem, L.O. Magnetic Resonance Imaging of Prostate Cancer. Clin. Imaging 2016, 40, 601–609. [Google Scholar] [CrossRef] [PubMed]
  26. Rahbar, H.; Partridge, S.C. Multiparametric Mr Imaging of Breast Cancer. Magn. Reson. Imaging Clin. N. Am. 2016, 24, 223–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Bourgeois, A.C.; Warren, L.A.; Chang, T.T.; Embry, S.; Hudson, K.; Bradley, Y.C. Role of Positron Emission Tomography/Computed Tomography in Breast Cancer. Radiol. Clin. N. Am. 2013, 51, 781–798. [Google Scholar] [CrossRef] [PubMed]
  28. Onal, C.; Findikcioglu, A.; Guler, O.C.; Reyhan, M.J. The Use of 18f-Fdg Positron Emission Tomography to Detect Mediastinal Lymph Nodes in Metastatic Breast Cancer. Breast 2020, 54, 197–202. [Google Scholar] [CrossRef]
  29. Shang, L.; Liu, Z.; Rong, Y.; Zhou, T.; Zuo, L.J. Mri in Breast Cancer Radiotherapy in Prone and Supine Positions. Front. Biosci. 2017, 22, 570–579. [Google Scholar]
  30. Bahreini, M.; Hosseinzadegan, A.; Rashidi, A.; Miri, S.R.; Mirzaei, H.R.; Hajian, P.J. A Raman-Based Serum Constituents’ Analysis for Gastric Cancer Diagnosis: In Vitro Study. Talanta 2019, 204, 826–832. [Google Scholar] [CrossRef]
  31. Caldarone, A.; Piccotti, F.; Morasso, C.; Truffi, M.; Sottotetti, F.; Guerra, C.; Albasini, S.; Agozzino, M.; Villani, L.; Corsi, F.; et al. Raman Analysis of Microcalcifications in Male Breast Cancer. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2021, 263, 120185. [Google Scholar] [CrossRef]
  32. Kopec, M.; Błaszczyk, M.; Radek, M.; Abramczyk, H.; Spectroscopy, B. Raman Imaging and Statistical Methods for Analysis Various Type of Human Brain Tumors and Breast Cancers. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2021, 262, 120091. [Google Scholar] [CrossRef]
  33. Auner, G.W.; Koya, S.K.; Huang, C.; Broadbent, B.; Trexler, M.; Auner, Z.; Elias, A.; Mehne, K.C.; Brusatori, M.A. Applications of Raman Spectroscopy in Cancer Diagnosis. Cancer Metastasis Rev. 2018, 37, 691–717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Paidi, S.K.; Shah, V.; Raj, P.; Glunde, K.; Pandey, R.; Barman, I.J. Coarse Raman and Optical Diffraction Tomographic Imaging Enable Label-Free Phenotyping of Isogenic Breast Cancer Cells of Varying Metastatic Potential. Biosens. Bioelectron. 2021, 175, 112863. [Google Scholar] [CrossRef] [PubMed]
  35. Fiorica, J.V. Breast Cancer Screening, Mammography, and Other Modalities. Clin. Obstet. Gynecol. 2016, 59, 688–709. [Google Scholar] [CrossRef] [PubMed]
  36. Gartlehner, G.; Thaler, K.J.; Chapman, A.; Kaminski, A.; Berzaczy, D.; Van Noord, M.G.; Helbich, T.H. Adjunct Ultrasonography for Breast Cancer Screening in Women At Average Risk: A Systematic Review. Int. J. Evid. Based Healthc. 2013, 11, 87–93. [Google Scholar] [CrossRef] [PubMed]
  37. Ramadan, S.Z. Methods Used in Computer-Aided Diagnosis for Breast Cancer Detection Using Mammograms: A Review. J. Healthc. Eng. 2020, 2020, 9162464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Sala, M.; Domingo, L.; Macià, F.; Comas, M.; Burón, A.; Castells, X. Does Digital Mammography Suppose An Advance in Early Diagnosis? Trends in Performance Indicators 6 Years After Digitalization. Eur. Radiol. 2015, 25, 850–859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Schettini, F.; Giuliano, M.; De Placido, S.; Arpino, G. Nab-Paclitaxel for the Treatment of Triple-Negative Breast Cancer: Rationale, Clinical Data and Future Perspectives. Cancer Treat. Rev. 2016, 50, 129–141. [Google Scholar] [CrossRef] [Green Version]
  40. Saadatmand, S.; Geuzinge, H.A.; Rutgers, E.J.; Mann, R.M.; Van Zuidewijn, D.B.D.R.; Zonderland, H.M.; Tollenaar, R.A.; Lobbes, M.B.; Ausems, M.G.; Van’t Riet, M. Mri Versus Mammography for Breast Cancer Screening in Women with Familial Risk (Famrisc): A Multicentre, Randomised, Controlled Trial. Lancet Oncol. 2019, 20, 1136–1147. [Google Scholar] [CrossRef] [Green Version]
  41. Jochelson, M. Contrast-Enhanced Digital Mammography. Radiol. Clin. 2014, 52, 609–616. [Google Scholar] [CrossRef] [PubMed]
  42. Lee-Felker, S.A.; Tekchandani, L.; Thomas, M.; Gupta, E.; Andrews-Tang, D.; Roth, A.; Sayre, J.; Rahbar, G. Newly Diagnosed Breast Cancer: Comparison of Contrast-Enhanced Spectral Mammography and Breast Mr Imaging in the Evaluation of Extent of Disease. Radiology 2017, 285, 389–400. [Google Scholar] [CrossRef] [Green Version]
  43. Mann, R.M.; Hooley, R.; Barr, R.G.; Moy, L. Novel Approaches to Screening for Breast Cancer. Radiology 2020, 297, 266–285. [Google Scholar] [CrossRef] [PubMed]
  44. Niikura, N.; Odisio, B.C.; Tokuda, Y.; Symmans, F.W.; Hortobagyi, G.N.; Ueno, N.T. Latest Biopsy Approach for Suspected Metastases in Patients with Breast Cancer. Nat. Rev. Clin. Oncol. 2013, 10, 711–719. [Google Scholar] [CrossRef] [PubMed]
  45. Tagliafico, A.S.; Piana, M.; Schenone, D.; Lai, R.; Massone, A.M.; Houssami, N.J. Overview of Radiomics in Breast Cancer Diagnosis and Prognostication. Breast 2020, 49, 74–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Tran, W.T.; Jerzak, K.; Lu, F.-I.; Klein, J.; Tabbarah, S.; Lagree, A.; Wu, T.; Rosado-Mendez, I.; Law, E.; Saednia, K.; et al. Personalized Breast Cancer Treatments Using Artificial Intelligence in Radiomics and Pathomics. J. Med. Imaging Radiat. Sci. 2019, 50, S32–S41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Forghani, R.; Savadjiev, P.; Chatterjee, A.; Muthukrishnan, N.; Reinhold, C.; Forghani, B. Radiomics and Artificial Intelligence for Biomarker and Prediction Model Development in Oncology. Comput. Struct. Biotechnol. J. 2019, 17, 995. [Google Scholar] [CrossRef]
  48. Gupta, R.; Kurc, T.; Sharma, A.; Almeida, J.S.; Saltz, J. The Emergence of Pathomics. Curr. Pathobiol. Rep. 2019, 7, 73–84. [Google Scholar] [CrossRef] [Green Version]
  49. Hendrix, N.; Hauber, B.; Lee, C.I.; Bansal, A.; Veenstra, D.L. Artificial Intelligence in Breast Cancer Screening: Primary Care Provider Preferences. J. Am. Med. Inform. Assoc. 2021, 28, 1117–1124. [Google Scholar] [CrossRef] [PubMed]
  50. Rodriguez-Ruiz, A.; Lång, K.; Gubern-Merida, A.; Broeders, M.; Gennaro, G.; Clauser, P.; Helbich, T.H.; Chevalier, M.; Tan, T.; Mertelmeier, T. Stand-Alone Artificial Intelligence for Breast Cancer Detection in Mammography: Comparison with 101 Radiologists. J. Nat. Cancer Inst. 2019, 111, 916–922. [Google Scholar] [CrossRef]
  51. Scimeca, M.; Urbano, N.; Toschi, N.; Bonanno, E.; Schillaci, O. Precision Medicine in Breast Cancer: From Biological Imaging to Artificial Intelligence. Semin. Cancer Biol. 2021, 72, 19. [Google Scholar] [CrossRef]
  52. Sechopoulos, I.; Teuwen, J.; Mann, R. Artificial Intelligence for Breast Cancer Detection in Mammography and Digital Breast Tomosynthesis: State of the Art. Semin. Cancer Biol. 2021, 72, 214–225. [Google Scholar] [CrossRef]
  53. Masaoutis, C.; Mihailidou, C.; Tsourouflis, G.; Theocharis, S. Exosomes in Lung Cancer Diagnosis and Treatment. From the Translating Research Into Future Clinical Practice. Biochimie 2018, 151, 27–36. [Google Scholar] [CrossRef] [PubMed]
  54. Wu, C.Y.; Du, S.L.; Zhang, J.; Liang, A.L.; Liu, Y.J. Exosomes and Breast Cancer: A Comprehensive Review of Novel Therapeutic Strategies from Diagnosis to Treatment. Cancer Gene Ther. 2017, 24, 6–12. [Google Scholar] [CrossRef] [PubMed]
  55. Zheng, Y.; Hasan, A.; Nejadi Babadaei, M.M.; Behzadi, E.; Nouri, M.; Sharifi, M.; Falahati, M. Exosomes: Multiple-Targeted Multifunctional Biological Nanoparticles in the Diagnosis, Drug Delivery, and Imaging of Cancer Cells. Biomed. Pharmacother. 2020, 129, 110442. [Google Scholar] [CrossRef] [PubMed]
  56. Hanjani, N.A.; Esmaelizad, N.; Zanganeh, S.; Gharavi, A.T.; Heidarizadeh, P.; Radfar, M.; Omidi, F.; Macloughlin, R.; Doroudian, M. Emerging Role of Exosomes As Biomarkers in Cancer Treatment and Diagnosis. Crit. Rev. Oncol. Hematol. 2022, 169, 103565. [Google Scholar] [CrossRef] [PubMed]
  57. Jafari, S.H.; Saadatpour, Z.; Salmaninejad, A.; Momeni, F.; Mokhtari, M.; Nahand, J.S.; Rahmati, M.; Mirzaei, H.; Kianmehr, M. Breast Cancer Diagnosis: Imaging Techniques and Biochemical Markers. J. Cell. Physiol. 2018, 233, 5200–5213. [Google Scholar] [CrossRef]
  58. Jia, Y.; Chen, Y.; Wang, Q.; Jayasinghe, U.; Luo, X.; Wei, Q.; Wang, J.; Xiong, H.; Chen, C.; Xu, B. Exosome: Emerging Biomarker in Breast Cancer. Oncotarget 2017, 8, 41717. [Google Scholar] [CrossRef] [Green Version]
  59. Li, G.; Hu, J.; Hu, G. Biomarker Studies in Early Detection and Prognosis of Breast Cancer. In Translational Research in Breast Cancer: Biomarker Diagnosis, Targeted Therapies and Approaches to Precision Medicine; Springer International Publishing: Cham, Switzerland, 2017; pp. 27–39. [Google Scholar] [CrossRef]
  60. Barzaman, K.; Moradi-Kalbolandi, S.; Hosseinzadeh, A.; Kazemi, M.H.; Khorramdelazad, H.; Safari, E.; Farahmand, L.J.I.I. Breast Cancer Immunotherapy: Current and Novel Approaches. Int. Immunopharmacol. 2021, 98, 107886. [Google Scholar] [CrossRef]
  61. Hristova, V.A.; Chan, D.W. Cancer Biomarker Discovery and Translation: Proteomics and Beyond. Expert Rev. Proteom. 2019, 16, 93–103. [Google Scholar] [CrossRef]
  62. Afzal, M.; Alharbi, K.S.; Alruwaili, N.K.; Al-Abassi, F.A.; Al-Malki, A.A.L.; Kazmi, I.; Kumar, V.; Kamal, M.A.; Nadeem, M.S.; Aslam, M.; et al. Nanomedicine in Treatment of Breast Cancer—A Challenge to Conventional Therapy. Semin. Cancer Biol. 2021, 69, 279–292. [Google Scholar] [CrossRef]
  63. Hashemzadeh, N.; Dolatkhah, M.; Adibkia, K.; Aghanejad, A.; Barzegar-Jalali, M.; Omidi, Y.; Barar, J. Recent Advances in Breast Cancer Immunotherapy: The Promising Impact of Nanomedicines. Life Sci. 2021, 271, 119110. [Google Scholar] [CrossRef]
  64. Boix-Montesinos, P.; Soriano-Teruel, P.M.; Armiñán, A.; Orzáez, M.; Vicent, M. The Past, Present, and Future of Breast Cancer Models for Nanomedicine Development. Adv. Drug Deliv. Rev. 2021, 173, 306–330. [Google Scholar] [CrossRef] [PubMed]
  65. Li, Q.; Dong, Z.; Chen, M.; Feng, L. Phenolic Molecules Constructed Nanomedicine for Innovative Cancer Treatment. Coord. Chem. Rev. 2021, 439, 213912. [Google Scholar] [CrossRef]
  66. Zhang, C.; Yan, L.; Wang, X.; Zhu, S.; Chen, C.; Gu, Z.; Zhao, Y. Progress, Challenges, and Future of Nanomedicine. Nano Today 2020, 35, 101008. [Google Scholar] [CrossRef]
  67. Nuzhina, J.V.; Shtil, A.A.; Prilepskii, A.Y.; Vinogradov, V.V. Preclinical Evaluation and Clinical Translation of Magnetite-Based Nanomedicines. J. Drug Deliv. Sci. Technol. 2019, 54, 101282. [Google Scholar] [CrossRef]
  68. Sousa, C.; Cruz, M.; Neto, A.; Pereira, K.; Peixoto, M.; Bastos, J.; Henriques, M.; Roda, D.; Marques, R.; Miranda, C.; et al. Neoadjuvant Radiotherapy in the Approach of Locally Advanced Breast Cancer. Ann. Oncol. 2020, 5, E000640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Montagna, G.; Ferraro, E.; Pilewskie, M.L. Neoadjuvant Chemotherapy for Nonmetastatic Breast Cancer: How Response Impacts Locoregional and Adjuvant Systemic Therapy Decision Making. Adv. Oncol. 2022, 2, 47–61. [Google Scholar] [CrossRef]
  70. Ataseven, B.; Von Minckwitz, G. The Impact of Neoadjuvant Treatment on Surgical Options and Outcomes. Ann. Surg. Oncol. 2016, 23, 3093–3099. [Google Scholar] [CrossRef]
  71. Dastjerd, N.T.; Valibeik, A.; Rahimi Monfared, S.; Goodarzi, G.; Moradi Sarabi, M.; Hajabdollahi, F.; Maniati, M.; Amri, J.; Tehrani, S.S. Gene Therapy: A Promising Approach for Breast Cancer Treatment. Cell Biochem. 2022, 40, 28–48. [Google Scholar] [CrossRef]
  72. Cao, K.; Abbassi, L.; Romano, E.; Kirova, Y. Radiation Therapy and Immunotherapy in Breast Cancer Treatment: Preliminary Data and Perspectives. Expert Rev. Anticancer Ther. 2021, 21, 501–510. [Google Scholar] [CrossRef]
  73. Orecchia, R. Radiation Therapy for Inflammatory Breast Cancer. Eur. J. Surg. Oncol. 2018, 44, 1148–1150. [Google Scholar] [CrossRef]
  74. García-Aranda, M.; Redondo, M. Immunotherapy: A Challenge of Breast Cancer Treatment. Cancers 2019, 11, 1822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Alexander, C.A.; Yang, Y.Y. Harnessing the Combined Potential of Cancer Immunotherapy and Nanomedicine: A New Paradigm in Cancer Treatment. Nanomed. Nanotechnol. Biol. Med. 2022, 40, 102492. [Google Scholar] [CrossRef] [PubMed]
  76. Amaoui, B.; Lalya, I.; Safini, F.; Semghouli, S. Combination of Immunotherapy-Radiotherapy in Non-Small Cell Lung Cancer: Reality and Perspective. Radiat. Med. Prot. 2021, 2, 160–164. [Google Scholar] [CrossRef]
  77. Jafarzadeh, L.; Khakpoor-Koosheh, M.; Mirzaei, H.; Mirzaei, H.R. Biomarkers for Predicting the Outcome of Various Cancer Immunotherapies. Crit. Rev. Oncol. Hematol. 2021, 157, 103161. [Google Scholar] [CrossRef] [PubMed]
  78. Disis, M.L.; Stanton, S.E. Immunotherapy in Breast Cancer: An Introduction. Breast 2018, 37, 196–199. [Google Scholar] [CrossRef] [Green Version]
  79. Skala, M.C.; Ayuso, J.M.; Burkard, M.E.; Deming, D.A. Breast Cancer Immunotherapy: Current Biomarkers and the Potential of in Vitro Assays. Curr. Opin. Biomed. Eng. 2022, 21, 100348. [Google Scholar] [CrossRef]
  80. Borgers, J.S.W.; Heimovaara, J.H.; Cardonick, E.; Dierickx, D.; Lambertini, M.; Haanen, J.B.A.G.; Amant, F. Immunotherapy for Cancer Treatment During Pregnancy. Lancet Oncol. 2021, 22, E550–E561. [Google Scholar] [CrossRef]
  81. Aumeeruddy, M.Z.; Mahomoodally, M.F. Combating Breast Cancer Using Combination Therapy with 3 Phytochemicals: Piperine, Sulforaphane, and Thymoquinone. Cancer 2019, 125, 1600–1611. [Google Scholar] [CrossRef]
  82. Morales, M.A.G.; Rodríguez, R.B.; Cruz, J.R.S.; Teran, L.M. Overview of New Treatments with Immunotherapy for Breast Cancer and A Proposal of A Combination Therapy. Molecules 2020, 25, 5686. [Google Scholar] [CrossRef]
  83. Núñez, C.; Capelo, J.L.; Igrejas, G.; Alfonso, A.; Botana, L.M.; Lodeiro, C. An Overview of the Effective Combination Therapies for the Treatment of Breast Cancer. Biomaterials 2016, 97, 34–50. [Google Scholar] [CrossRef]
  84. Ma, J.; Li, X.; Zhang, Q.; Li, N.; Sun, S.; Zhao, S.; Zhao, Z.; Li, M. A Novel Treatment Strategy of Her2-Targeted Therapy in Combination with Everolimus for Hr+/Her2- Advanced Breast Cancer Patients with Her2 Mutations. Transl. Oncol. 2022, 21, 101444. [Google Scholar] [CrossRef]
  85. Ahmed, M.; Moussa, M.; Goldberg, S.N. Synergy in Cancer Treatment Between Liposomal Chemotherapeutics and Thermal Ablation. Chem. Phy. Lipids 2012, 165, 424–437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Poel, H.G.; Bergh, R.C.N.; Briers, E.; Cornford, P.; Govorov, A.; Henry, A.M.; Lam, T.B.; Mason, M.D.; Rouvière, O.; Santis, M.D.; et al. Focal Therapy in Primary Localised Prostate Cancer: The European Association of Urology Position in 2018. Eur. Urol. 2018, 74, 84–91. [Google Scholar] [CrossRef] [PubMed]
  87. Vogl, T.J.; Farshid, P.; Naguib, N.N.N.; Zangos, S. Thermal Ablation Therapies in Patients with Breast Cancer Liver Metastases: A Review. Eur. Radiol. 2013, 23, 797–804. [Google Scholar] [CrossRef] [PubMed]
  88. Voort, E.M.; Struik, G.M.; Birnie, E.; Moelker, A.; Verhoef, C.; Klem, T.M. Thermal Ablation As An Alternative for Surgical Resection of Small (≤2 cm) Breast Cancers: A Meta-Analysis. Clin. Breast Cancer 2021, 21, E715–E730. [Google Scholar] [CrossRef] [PubMed]
  89. Barral, M.; Auperin, A.; Hakime, A.; Cartier, V.; Tacher, V.; Otmezguine, Y.; Tselikas, L.; De Baere, T.; Deschamps, F. Percutaneous Thermal Ablation of Breast Cancer Metastases in Oligometastatic Patients. Cardiovasc. Interv. Radiol. 2016, 39, 885–893. [Google Scholar] [CrossRef] [PubMed]
  90. Pusceddu, C.; Paliogiannis, P.; Nigri, G.; Fancellu, A. Cryoablation in the Management of Breast Cancer: Evidence to Date. Breast Cancer: Targets Ther. 2019, 11, 283. [Google Scholar] [CrossRef] [Green Version]
  91. Roubidoux, M.A.; Yang, W.; Stafford, R. Image-Guided Ablation in Breast Cancer Treatment. Tech. Vasc. Interv. Radiol. 2014, 17, 49–54. [Google Scholar] [CrossRef]
  92. Webb, H.; Lubner, M.G.; Hinshaw, J.L. Thermal Ablation. Semin. Roentgenol. 2011, 46, 133–141. [Google Scholar] [CrossRef]
  93. Chatterjee, A.; He, D.; Fan, X.; Wang, S.; Szasz, T.; Yousuf, A.; Pineda, F.; Antic, T.; Mathew, M.; Karczmar, G.S.; et al. Performance of Ultrafast Dce-Mri for Diagnosis of Prostate Cancer. Acad. Radiol. 2018, 25, 349–358. [Google Scholar] [CrossRef]
  94. Peiravi, M.; Eslami, H.; Ansari, M.; Zare-Zardini, H. Magnetic Hyperthermia: Potentials and Limitations. J. Ind. Chem. Soc. 2022, 99, 100269. [Google Scholar] [CrossRef]
  95. Jose, J.; Kumar, R.; Harilal, S.; Mathew, G.E.; Parambi, D.G.T.; Prabhu, A.; Uddin, M.; Aleya, L.; Kim, H.; Mathew, B. Magnetic Nanoparticles for Hyperthermia in Cancer Treatment: An Emerging Tool. Environ. Sci. Pollut. Res. 2020, 27, 19214–19225. [Google Scholar] [CrossRef] [PubMed]
  96. Ezzati, M.; Yousefi, B.; Velaei, K.; Safa, A. A Review on Anti-Cancer Properties of Quercetin in Breast Cancer. Life Sci. 2020, 248, 117463. [Google Scholar] [CrossRef] [PubMed]
  97. Binienda, A.; Ziolkowska, S.; Pluciennik, E. The Anticancer Properties of Silibinin: Its Molecular Mechanism and Therapeutic Effect in Breast Cancer. Anticancer Agents Med. Chem. 2020, 20, 1787–1796. [Google Scholar] [CrossRef]
  98. Bonofiglio, D.; Giordano, C.; Francesca, A.; Lanzino, M.; Sebastiano, A. Natural Products As Promising Antitumoral Agents in Breast Cancer: Mechanisms of Action and Molecular Targets. Mini Rev. Med. Chem. 2016, 16, 596–604. [Google Scholar] [CrossRef]
  99. Fasoulakis, Z.; Koutras, A.; Syllaios, A.; Schizas, D.; Garmpis, N.; Diakosavvas, M.; Angelou, K.; Tsatsaris, G.; Pagkalos, A.; Ntounis, T.; et al. Breast Cancer Apoptosis and the Therapeutic Role of Luteolin. Chirurgia 2021, 116, 170–177. [Google Scholar] [CrossRef]
  100. Cheuk, I.W.; Chen, J.; Siu, M.; Ho, J.C.; Lam, S.S.; Shin, V.Y.; Kwong, A.A. Resveratrol Enhanced Chemosensitivity by Reversing Macrophage Polarization in Breast Cancer. Clin. Transl. Oncol. 2022, 24, 854–863. [Google Scholar] [CrossRef]
  101. Sharma, P.; Mcclees, S.F.; Afaq, F. Pomegranate for Prevention and Treatment of Cancer: An Update. Molecules 2017, 22, 177. [Google Scholar] [CrossRef] [Green Version]
  102. Rappuoli, R.; Pizza, M.; Del Giudice, G.; De Gregorio, E. Vaccines, New Opportunities for A New Society. Proc. Natl. Acad. Sci. USA 2014, 111, 12288–12293. [Google Scholar] [CrossRef] [Green Version]
  103. Igarashi, Y.; Sasada, T. Cancer Vaccines: Toward the Next Breakthrough in Cancer Immunotherapy. J. Immunol. Res. 2020, 2020, 5825401. [Google Scholar] [CrossRef]
  104. Smith, R.T. Immune Surveillance; Elsevier: Amsterdam, The Netherlands, 2012. [Google Scholar]
  105. Bitton, R.J. Cancer Vaccines: A Critical Review on Clinical Impact. Curr. Opin. Mol. Ther. 2004, 6, 17–26. [Google Scholar] [PubMed]
  106. Starling, S. Immune Editing Shapes the Cancer Landscape. Nat. Rev. Immunol. 2017, 17, 729. [Google Scholar] [CrossRef] [PubMed]
  107. Hegde, P.S.; Chen, D.S. Top 10 Challenges in Cancer Immunotherapy. Immunity 2020, 52, 17–35. [Google Scholar] [CrossRef]
  108. Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A Guide to Cancer Immunotherapy: From T Cell Basic Science to Clinical Practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef] [PubMed]
  109. Mccarthy, E.F. The Toxins of William B. Coley and the Treatment of Bone and Soft-Tissue Sarcomas. Iowa Orthop. J. 2006, 26, 154–158. [Google Scholar]
  110. Grimmett, E.; Al-Share, B.; Alkassab, M.B.; Zhou, R.W.; Desai, A.; Rahim, M.M.A.; Woldie, I. Cancer Vaccines: Past, Present and Future; A Review Article. Discov. Oncol. 2022, 13, 31. [Google Scholar] [CrossRef]
  111. Sela, M.; Mozes, E. Therapeutic Vaccines in Autoimmunity. Proc. Natl. Acad. Sci. USA 2004, 101 (Suppl. S2), 14586–14592. [Google Scholar] [CrossRef] [Green Version]
  112. Guo, C.; Manjili, M.H.; Subjeck, J.R.; Sarkar, D.; Fisher, P.B.; Wang, X.Y. Therapeutic Cancer Vaccines: Past, Present, and Future. Adv. Cancer Res. 2013, 119, 421–475. [Google Scholar] [CrossRef] [Green Version]
  113. Pan, C.; Yue, H.; Zhu, L.; Ma, G.-H.; Wang, H.-L. Prophylactic Vaccine Delivery Systems Against Epidemic Infectious Diseases. Adv. Drug Deliv. Rev. 2021, 176, 113867. [Google Scholar] [CrossRef]
  114. Burke, E.E.; Kodumudi, K.; Ramamoorthi, G.; Czerniecki, B.J. Vaccine Therapies for Breast Cancer. Surg. Oncol. Clin. 2019, 28, 353–367. [Google Scholar] [CrossRef]
  115. Stern, P.L. Key Steps in Vaccine Development. Ann. Allergy Asthma Immunol. 2020, 125, 17–27. [Google Scholar] [CrossRef]
  116. Desai, R.; Coxon, A.T.; Dunn, G.P. Therapeutic Applications of the Cancer Immunoediting Hypothesis. Semin. Cancer Biol. 2022, 78, 63–77. [Google Scholar] [CrossRef] [PubMed]
  117. Alves-Fernandes, D.K.; Jasiulionis, M.G. The Role of Sirt1 on Dna Damage Response and Epigenetic Alterations in Cancer. Int. J. Mol. Sci. 2019, 20, 3153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Saito, E.; Kuo, R.; Kramer, K.R.; Gohel, N.; Giles, D.A.; Moore, B.B.; Miller, S.D.; Shea, L.D. Design of Biodegradable Nanoparticles to Modulate Phenotypes of Antigen-Presenting Cells for Antigen-Specific Treatment of Autoimmune Disease. Biomaterials 2019, 222, 119432. [Google Scholar] [CrossRef] [PubMed]
  119. Shaaban, M.; Othman, H.; Ibrahim, T.; Ali, M.; Abdelmoaty, M.; Abdel-Kawi, A.-R.; Mostafa, A.; El Nakeeb, A.; Emam, H.; Refaat, A. Immune Checkpoint Regulators: A New Era Toward Promising Cancer Therapy. Curr. Cancer Drug Targets 2020, 20, 429–460. [Google Scholar] [CrossRef]
  120. Dersh, D.; Hollý, J.; Yewdell, J.W. A Few Good Peptides: Mhc Class I-Based Cancer Immunosurveillance and Immunoevasion. Nat. Rev. Immunol. 2021, 21, 116–128. [Google Scholar] [CrossRef]
  121. Johnson, A.M.; Bullock, B.L.; Neuwelt, A.J.; Poczobutt, J.M.; Kaspar, R.E.; Li, H.Y.; Kwak, J.W.; Hopp, K.; Weiser-Evans, M.C.; Heasley, L.E. Cancer Cell–Intrinsic Expression of Mhc Class Ii Regulates the Immune Microenvironment and Response to Anti–Pd-1 Therapy in Lung Adenocarcinoma. J. Immunol. 2020, 204, 2295–2307. [Google Scholar] [CrossRef]
  122. Criscitiello, C. Tumor-Associated Antigens in Breast Cancer. Breast Care 2012, 7, 262–266. [Google Scholar] [CrossRef] [Green Version]
  123. Fracol, M.; Shah, N.; Dolivo, D.; Hong, S.; Giragosian, L.; Galiano, R.; Mustoe, T.; Kim, J. Can Breast Implants Induce Breast Cancer Immunosurveillance? An Analysis of Antibody Response to Breast Cancer Antigen Following Implant Placement. Plast. Reconstr. Surg. 2021, 148, 287–298. [Google Scholar] [CrossRef]
  124. Huber, V.; Camisaschi, C.; Berzi, A.; Ferro, S.; Lugini, L.; Triulzi, T.; Tuccitto, A.; Tagliabue, E.; Castelli, C.; Rivoltini, L. Cancer Acidity: An Ultimate Frontier of Tumor Immune Escape and A Novel Target of Immunomodulation. Semin. Cancer Biol. 2017, 43, 74–89. [Google Scholar] [CrossRef]
  125. Shimizu, K.; Iyoda, T.; Okada, M.; Yamasaki, S.; Fujii, S.I. Immune Suppression and Reversal of the Suppressive Tumor Microenvironment. Int. Immunol. 2018, 30, 445–454. [Google Scholar] [CrossRef] [PubMed]
  126. Wan, Y.Y.; Flavell, R.A. ‘Yin-Yang’ Functions of Transforming Growth Factor-Beta and T Regulatory Cells in Immune Regulation. Immunol. Rev. 2007, 220, 199–213. [Google Scholar] [CrossRef] [PubMed]
  127. Cornel, A.M.; Mimpen, I.L.; Nierkens, S. Mhc Class I Downregulation in Cancer: Underlying Mechanisms and Potential Targets for Cancer Immunotherapy. Cancers 2020, 12, 1760. [Google Scholar] [CrossRef] [PubMed]
  128. Guerrouahen, B.S.; Maccalli, C.; Cugno, C.; Rutella, S.; Akporiaye, E.T. Reverting Immune Suppression to Enhance Cancer Immunotherapy. Front. Oncol. 2020, 9, 1554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Oshi, M.; Asaoka, M.; Tokumaru, Y.; Angarita, F.A.; Yan, L.; Matsuyama, R.; Zsiros, E.; Ishikawa, T.; Endo, I.; Takabe, K. Abundance of Regulatory T Cell (Treg) As A Predictive Biomarker for Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer. Cancers 2020, 12, 3038. [Google Scholar] [CrossRef]
  130. Fasoulakis, Z.; Kolios, G.; Papamanolis, V.; Kontomanolis, E.N. Interleukins Associated with Breast Cancer. Cureus 2018, 10, E3549. [Google Scholar] [CrossRef] [Green Version]
  131. Van Den Hove, L.E.; Vandenberghe, P.; Van Gool, S.W.; Ceuppens, J.L.; Demuynck, H.; Verhoef, G.E.; Boogaerts, M.A. Peripheral Blood Lymphocyte Subset Shifts in Patients with Untreated Hematological Tumors: Evidence for Systemic Activation of the T Cell Compartment. Leuk. Res. 1998, 22, 175–184. [Google Scholar] [CrossRef]
  132. Schütz, F.; Stefanovic, S.; Mayer, L.; Von Au, A.; Domschke, C.; Sohn, C. Pd-1/Pd-L1 Pathway in Breast Cancer. Oncol. Res. Treat. 2017, 40, 294–297. [Google Scholar] [CrossRef]
  133. Planes-Laine, G.; Rochigneux, P.; Bertucci, F.; Chrétien, A.S.; Viens, P.; Sabatier, R.; Gonçalves, A. Pd-1/Pd-L1 Targeting in Breast Cancer: The First Clinical Evidences Are Emerging. A Literature Review. Cancers 2019, 11, 1033. [Google Scholar] [CrossRef] [Green Version]
  134. Lü, M.-H.; Liao, Z.-L.; Zhao, X.-Y.; Fan, Y.-H.; Lin, X.-L.; Fang, D.-C.; Guo, H.; Yang, S.-M. Htert-Based Therapy: A Universal Anticancer Approach. Oncol. Rep. 2012, 28, 1945–1952. [Google Scholar] [CrossRef] [Green Version]
  135. Krishnamurti, U.; Silverman, J.F. Her2 in Breast Cancer: A Review and Update. Adv. Anat. Pathol. 2014, 21, 100–107. [Google Scholar] [CrossRef] [PubMed]
  136. Swain, S.M.; Shastry, M.; Hamilton, E. Targeting Her2-Positive Breast Cancer: Advances and Future Directions. Nat. Rev. Drug Discov. 2022, 22, 101–126. [Google Scholar] [CrossRef] [PubMed]
  137. Martínez-Sáez, O.; Prat, A. Current and Future Management of Her2-Positive Metastatic Breast Cancer. J. Oncol. Pract. 2021, 17, 594–604. [Google Scholar] [CrossRef] [PubMed]
  138. Mitri, Z.; Constantine, T.; O’regan, R. The Her2 Receptor in Breast Cancer: Pathophysiology, Clinical Use, and New Advances in Therapy. Chemother. Res. Pract. 2012, 2012, 743193. [Google Scholar] [CrossRef] [Green Version]
  139. Yang, P.; Du, C.W.; Kwan, M.; Liang, S.X.; Zhang, G.J. The Impact of P53 in Predicting Clinical Outcome of Breast Cancer Patients with Visceral Metastasis. Sci. Rep. 2013, 3, 2246. [Google Scholar] [CrossRef] [Green Version]
  140. Berke, T.P.; Slight, S.H.; Hyder, S.M. Role of Reactivating Mutant P53 Protein in Suppressing Growth and Metastasis of Triple-Negative Breast Cancer. Onco Targets Ther. 2022, 15, 23. [Google Scholar] [CrossRef]
  141. Gasco, M.; Shami, S.; Crook, T. The P53 Pathway in Breast Cancer. Breast Cancer Res. 2002, 4, 70–76. [Google Scholar] [CrossRef] [Green Version]
  142. Duffy, M.J.; Synnott, N.C.; Crown, J. Mutant P53 in Breast Cancer: Potential As A Therapeutic Target and Biomarker. Breast Cancer Res. Treat. 2018, 170, 213–219. [Google Scholar] [CrossRef]
  143. Kufe, D.W. Muc1-C Oncoprotein As A Target in Breast Cancer: Activation of Signaling Pathways and Therapeutic Approaches. Oncogene 2013, 32, 1073–1081. [Google Scholar] [CrossRef] [Green Version]
  144. Zaretsky, J.Z.; Barnea, I.; Aylon, Y.; Gorivodsky, M.; Wreschner, D.H.; Keydar, I. Muc1 Gene Overexpressed in Breast Cancer: Structure and Transcriptional Activity of the Muc1 Promoter and Role of Estrogen Receptor Alpha (Erα) in Regulation of the Muc1 Gene Expression. Mol. Cancer 2006, 5, 57. [Google Scholar] [CrossRef] [Green Version]
  145. Taylor-Papadimitriou, J.; Burchell, J.; Miles, D.W.; Dalziel, M. Muc1 and Cancer. Biochim. Biophys. Acta Mol. Basis Dis. 1999, 1455, 301–313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Guillen-Poza, P.A.; Sánchez-Fernández, E.M.; Artigas, G.; García Fernández, J.M.; Hinou, H.; Ortiz Mellet, C.; Nishimura, S.-I.; Garcia-Martin, F. Amplified Detection of Breast Cancer Autoantibodies Using Muc1-Based Tn Antigen Mimics. J. Med. Chem. 2020, 63, 8524–8533. [Google Scholar] [CrossRef] [PubMed]
  147. Anoop, T.M.; Joseph, P.R.; Soman, S.; Chacko, S.; Mathew, M. Significance of Serum Carcinoembryonic Antigen in Metastatic Breast Cancer Patients: A Prospective Study. World J. Clin. Oncol. 2022, 13, 529–539. [Google Scholar] [CrossRef] [PubMed]
  148. Kabel, A.M. Tumor Markers of Breast Cancer: New Prospectives. J. Oncol. Sci. 2017, 3, 5–11. [Google Scholar] [CrossRef]
  149. Imamura, M.; Morimoto, T.; Nomura, T.; Michishita, S.; Nishimukai, A.; Higuchi, T.; Fujimoto, Y.; Miyagawa, Y.; Kira, A.; Murase, K.; et al. Independent Prognostic Impact of Preoperative Serum Carcinoembryonic Antigen and Cancer Antigen 15-3 Levels for Early Breast Cancer Subtypes. World J. Surg. Oncol. 2018, 16, 26. [Google Scholar] [CrossRef] [Green Version]
  150. Kirkpatrick, K.L.; Ogunkolade, W.; Elkak, A.E.; Bustin, S.; Jenkins, P.; Ghilchick, M.; Newbold, R.F.; Mokbel, K. Htert Expression in Human Breast Cancer and Non-Cancerous Breast Tissue: Correlation with Tumour Stage and C-Myc Expression. Breast Cancer Res. Treat. 2003, 77, 277–284. [Google Scholar] [CrossRef]
  151. Dratwa, M.; Wysoczańska, B.; Łacina, P.; Kubik, T.; Bogunia-Kubik, K. Tert—Regulation and Roles in Cancer Formation. Front. Immunol. 2020, 11, 589929. [Google Scholar] [CrossRef]
  152. Kirkpatrick, K.L.; Clark, G.; Ghilchick, M.; Newbold, R.F.; Mokbel, K. Htert Mrna Expression Correlates with Telomerase Activity in Human Breast Cancer. Eur. J. Surg. Oncol. 2003, 29, 321–326. [Google Scholar] [CrossRef] [Green Version]
  153. Davodabadi, F.; Sarhadi, M.; Arabpour, J.; Sargazi, S.; Rahdar, A.; Díez-Pascual, A.M. Breast Cancer Vaccines: New Insights Into Immunomodulatory and Nano-Therapeutic Approaches. J. Control. Release 2022, 349, 844–875. [Google Scholar] [CrossRef]
  154. Pallerla, S.; Abdul, A.U.R.M.; Comeau, J.; Jois, S. Cancer Vaccines, Treatment of the Future: With Emphasis on Her2-Positive Breast Cancer. Int. J. Mol. Sci. 2021, 22, 779. [Google Scholar] [CrossRef]
  155. Koido, S. Dendritic-Tumor Fusion Cell-Based Cancer Vaccines. Int. J. Mol. Sci. 2016, 17, 828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Cuzzubbo, S.; Mangsbo, S.; Nagarajan, D.; Habra, K.; Pockley, A.G.; Mcardle, S.E.B. Cancer Vaccines: Adjuvant Potency, Importance of Age, Lifestyle, and Treatments. Front. Immunol. 2021, 11, 615240. [Google Scholar] [CrossRef] [PubMed]
  157. Zhu, S.-Y.; Yu, K.-D. Breast Cancer Vaccines: Disappointing Or Promising? Front. Immunol. 2022, 13, 828386. [Google Scholar] [CrossRef]
  158. De Paula Peres, L.; Da Luz, F.A.C.; Dos Anjos Pultz, B.; Brígido, P.C.; De Araújo, R.A.; Goulart, L.R.; Silva, M.J.B. Peptide Vaccines in Breast Cancer: The Immunological Basis for Clinical Response. Biotechnol. Adv. 2015, 33, 1868–1877. [Google Scholar] [CrossRef] [PubMed]
  159. Wang, L.; Wang, N.; Zhang, W.; Cheng, X.; Yan, Z.; Shao, G.; Wang, X.; Wang, R.; Fu, C. Therapeutic Peptides: Current Applications and Future Directions. Signal Transduct. Target. Ther. 2022, 7, 48. [Google Scholar] [CrossRef]
  160. Nicolás-Morales, M.L.; Luisa-Sanjuan, A.; Gutiérrez-Torres, M.; Vences-Velázquez, A.; Ortuño-Pineda, C.; Espinoza-Rojo, M.; Navarro-Tito, N.; Cortés-Sarabia, K. Peptide-Based Vaccines in Clinical Phases and New Potential Therapeutic Targets As A New Approach for Breast Cancer: A Review. Vaccines 2022, 10, 1249. [Google Scholar] [CrossRef]
  161. Liu, J.; Fu, M.; Wang, M.; Wan, D.; Wei, Y.; Wei, X. Cancer Vaccines As Promising Immuno-Therapeutics: Platforms and Current Progress. J. Hematol. Oncol. 2022, 15, 28. [Google Scholar] [CrossRef]
  162. Corti, C.; Giachetti, P.P.M.B.; Eggermont, A.M.M.; Delaloge, S.; Curigliano, G. Therapeutic Vaccines for Breast Cancer: Has the Time Finally Come? Eur. J. Cancer 2022, 160, 150–174. [Google Scholar] [CrossRef]
  163. Kang, J.; Lee, H.-J.; Lee, J.; Hong, J.; Hong Kim, Y.; Disis, M.L.; Gim, J.-A.; Park, K.H. Novel Peptide-Based Vaccine Targeting Heat Shock Protein 90 Induces Effective Antitumor Immunity in A Her2+ Breast Cancer Murine Model. J. Immunother. Cancer 2022, 10, E004702. [Google Scholar] [CrossRef]
  164. Eavarone, D.A.; Al-Alem, L.; Lugovskoy, A.; Prendergast, J.M.; Nazer, R.I.; Stein, J.N.; Dransfield, D.T.; Behrens, J.; Rueda, B.R. Humanized Anti-Sialyl-Tn Antibodies for the Treatment of Ovarian Carcinoma. PLoS ONE 2018, 13, E0201314. [Google Scholar] [CrossRef]
  165. Munkley, J. The Role of Sialyl-Tn in Cancer. Int. Mol. Sci. 2016, 17, 275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Vorup-Jensen, T. On the Roles of Polyvalent Binding in Immune Recognition: Perspectives in the Nanoscience of Immunology and the Immune Response to Nanomedicines. Adv. Drug Deliv. Rev. 2012, 64, 1759–1781. [Google Scholar] [CrossRef] [PubMed]
  167. Ward, S.; Casey, D.; Labarthe, M.-C.; Whelan, M.; Dalgleish, A.; Pandha, H.; Todryk, S. Immunotherapeutic Potential of Whole Tumour Cells. Cancer Immunol. Immunother. 2002, 51, 351–357. [Google Scholar] [CrossRef] [PubMed]
  168. Al-Awadhi, A.; Lee Murray, J.; Ibrahim, N.K. Developing Anti-Her2 Vaccines: Breast Cancer Experience. Int. J. Cancer 2018, 143, 2126–2132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Leitner, W.W.; Ying, H.; Restifo, N.P. Dna and Rna-Based Vaccines: Principles, Progress and Prospects. Vaccine 1999, 18, 765–777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Fritah, H.; Rovelli, R.; Chiang, C.L.-L.; Kandalaft, L.E. The Current Clinical Landscape of Personalized Cancer Vaccines. Cancer Treat. Rev. 2022, 106, 102383. [Google Scholar] [CrossRef]
  171. Bird, R.C.; Deinnocentes, P.; Church Bird, A.E.; Lutful Kabir, F.M.; Martinez-Romero, E.G.; Smith, A.N.; Smith, B.F. Autologous Hybrid Cell Fusion Vaccine in A Spontaneous Intermediate Model of Breast Carcinoma. J. Vet. Sci. 2019, 20, E48. [Google Scholar] [CrossRef]
  172. Paston, S.J.; Brentville, V.A.; Symonds, P.; Durrant, L.G. Cancer Vaccines, Adjuvants, and Delivery Systems. Front. Immunol. 2021, 12, 627932. [Google Scholar] [CrossRef]
  173. Khong, H.; Overwijk, W.W. Adjuvants for Peptide-Based Cancer Vaccines. J. Immunother. Cancer 2016, 4, 56. [Google Scholar] [CrossRef] [Green Version]
  174. Dubensky, T.W.; Reed, S.G. Adjuvants for Cancer Vaccines. Semin. Immunol. 2010, 22, 155–161. [Google Scholar] [CrossRef]
  175. He, X.; Zhou, S.; Huang, W.-C.; Seffouh, A.; Mabrouk, M.T.; Morgan, M.T.; Ortega, J.; Abrams, S.I.; Lovell, J.F. A Potent Cancer Vaccine Adjuvant System for Particleization of Short, Synthetic Cd8+ T Cell Epitopes. Acs Nano 2021, 15, 4357–4371. [Google Scholar] [CrossRef] [PubMed]
  176. Bobanga, I.D.; Petrosiute, A.; Huang, A.Y. Chemokines As Cancer Vaccine Adjuvants. Vaccines 2013, 1, 444–462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Knutson, K.L.; Schiffman, K.; Disis, M.L. Immunization with A Her-2/Neu Helper Peptide Vaccine Generates Her-2/Neu Cd8 T-Cell Immunity in Cancer Patients. J. Clin. Investig. 2001, 107, 477–484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Malonis, R.J.; Lai, J.R.; Vergnolle, O. Peptide-Based Vaccines: Current Progress and Future Challenges. Chem. Rev. 2020, 120, 3210–3229. [Google Scholar] [CrossRef] [Green Version]
  179. Tobias, J.; Garner-Spitzer, E.; Drinić, M.; Wiedermann, U. Vaccination Against Her-2/Neu, with Focus on Peptide-Based Vaccines. Ann. Oncol. 2022, 7, 100361. [Google Scholar] [CrossRef] [PubMed]
  180. Nordin, M.L.; Mohamad Norpi, A.S.; Ng, P.Y.; Yusoff, K.; Abu, N.; Lim, K.P.; Azmi, F. Her2/Neu-Based Peptide Vaccination-Pulsed with B-Cell Epitope Induced Efficient Prophylactic and Therapeutic Antitumor Activities in Tubo Breast Cancer Mice Model. Cancers 2021, 13, 4958. [Google Scholar] [CrossRef]
  181. Pierini, S.; Perales-Linares, R.; Uribe-Herranz, M.; Pol, J.G.; Zitvogel, L.; Kroemer, G.; Facciabene, A.; Galluzzi, L. Trial Watch: Dna-Based Vaccines for Oncological Indications. Oncoimmunology 2017, 6, E1398878. [Google Scholar] [CrossRef]
  182. Benedetti, R.; Dell’aversana, C.; Giorgio, C.; Astorri, R.; Altucci, L. Breast Cancer Vaccines: New Insights. Front. Endocrinol. 2017, 8, 270. [Google Scholar] [CrossRef] [Green Version]
  183. Holmberg, L.A.; Sandmaier, B.M. Theratope® Vaccine (Stn-Klh). Expert Opin. Biol. Ther. 2001, 1, 881–891. [Google Scholar] [CrossRef]
  184. Kwa, M.J.; Adams, S. Checkpoint Inhibitors in Triple-Negative Breast Cancer (Tnbc): Where to Go from Here. Cancer 2018, 124, 2086–2103. [Google Scholar] [CrossRef] [Green Version]
  185. Emens, L.A.; Esteva, F.J.; Beresford, M.; Saura, C.; De Laurentiis, M.; Kim, S.-B.; Im, S.-A.; Wang, Y.; Salgado, R.; Mani, A.; et al. Trastuzumab Emtansine Plus Atezolizumab Versus Trastuzumab Emtansine Plus Placebo in Previously Treated, Her2-Positive Advanced Breast Cancer (Kate2): A Phase 2, Multicentre, Randomised, Double-Blind Trial. Lancet Oncol. 2020, 21, 1283–1295. [Google Scholar] [CrossRef] [PubMed]
  186. Fares, J.; Kanojia, D.; Rashidi, A.; Ulasov, I.; Lesniak, M.S. Landscape of Combination Therapy Trials in Breast Cancer Brain Metastasis. Int. J. Cancer 2020, 147, 1939–1952. [Google Scholar] [CrossRef] [PubMed]
  187. Fujiwara, Y.; Sun, Y.; Torphy, R.J.; He, J.; Yanaga, K.; Edil, B.H.; Schulick, R.D.; Zhu, Y. Pomalidomide Inhibits Pd-L1 Induction to Promote Antitumor Immunity. Cancer Res. 2018, 78, 6655–6665. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Lai, J.; Beavis, P.A.; Li, J.; Darcy, P.K. Augmenting Adoptive T-Cell Immunotherapy by Targeting the Pd-1/Pd-L1 Axis. Cancer Res. 2021, 81, 5803–5805. [Google Scholar] [CrossRef]
  189. Roy, S.; Sethi, T.K.; Taylor, D.; Kim, Y.J.; Johnson, D.B. Breakthrough Concepts in Immune-Oncology: Cancer Vaccines At the Bedside. J. Leuk. Biol. 2020, 108, 1455–1489. [Google Scholar] [CrossRef]
  190. Crosby, E.J.; Acharya, C.R.; Haddad, A.-F.; Rabiola, C.A.; Lei, G.; Wei, J.-P.; Yang, X.-Y.; Wang, T.; Liu, C.-X.; Wagner, K.U.; et al. Stimulation of Oncogene-Specific Tumor-Infiltrating T Cells Through Combined Vaccine and Apd-1 Enable Sustained Antitumor Responses Against Established Her2 Breast Cancer. Clin. Cancer Res. 2020, 26, 4670–4681. [Google Scholar] [CrossRef] [PubMed]
  191. Gall, V.A.; Philips, A.V.; Qiao, N.; Clise-Dwyer, K.; Perakis, A.A.; Zhang, M.; Clifton, G.T.; Sukhumalchandra, P.; Ma, Q.; Reddy, S.M.; et al. Trastuzumab Increases Her2 Uptake and Cross-Presentation by Dendritic Cells. Cancer Res. 2017, 77, 5374–5383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Correia, A.S.; Gärtner, F.; Vale, N. Drug Combination and Repurposing for Cancer Therapy: The Example of Breast Cancer. Heliyon 2021, 7, E05948. [Google Scholar] [CrossRef]
  193. Hodge, J.W.; Ardiani, A.; Farsaci, B.; Kwilas, A.R.; Gameiro, S.R. The Tipping Point for Combination Therapy: Cancer Vaccines with Radiation, Chemotherapy, Or Targeted Small Molecule Inhibitors. Semin. Oncol. 2012, 39, 323–339. [Google Scholar] [CrossRef] [Green Version]
  194. Nicolini, A.; Carpi, A.; Ferrari, P.; Mario Biava, P.; Rossi, G. Immunotherapy and Hormone-Therapy in Metastatic Breast Cancer: A Review and An Update. Curr. Drug Targets 2016, 17, 1127–1139. [Google Scholar] [CrossRef]
  195. Sertoli, M.R.; Scarsi, P.G.; Rosso, R. Rationale for Combining Chemotherapy and Hormonal Therapy in Breast Cancer. J. Steroid Biochem. 1985, 23, 1097–1103. [Google Scholar] [CrossRef]
  196. Mohit, E.; Hashemi, A.; Allahyari, M. Breast Cancer Immunotherapy: Monoclonal Antibodies and Peptide-Based Vaccines. Expert Rev. Clin. Immunol. 2014, 10, 927–961. [Google Scholar] [CrossRef] [PubMed]
  197. Wolfson, B.; Franks, S.E.; Hodge, J.W. Stay on Target: Reengaging Cancer Vaccines in Combination Immunotherapy. Vaccines 2021, 9, 509. [Google Scholar] [CrossRef] [PubMed]
  198. Mirjolet, C.; Truc, G. [Abscopal Effect: Myth Or Reality?]. Cancer Radiother. 2021, 25, 533–536. [Google Scholar] [CrossRef] [PubMed]
  199. Sindoni, A.; Minutoli, F.; Ascenti, G.; Pergolizzi, S. Combination of Immune Checkpoint Inhibitors and Radiotherapy: Review of the Literature. Crit. Rev. Oncol. Hematol. 2017, 113, 63–70. [Google Scholar] [CrossRef]
  200. Antonarelli, G.; Corti, C.; Tarantino, P.; Ascione, L.; Cortes, J.; Romero, P.; Mittendorf, E.A.; Disis, M.L.; Curigliano, G. Therapeutic Cancer Vaccines Revamping: Technology Advancements and Pitfalls. Ann. Oncol. 2021, 32, 1537–1551. [Google Scholar] [CrossRef]
  201. Li, W.; Joshi, M.D.; Singhania, S.; Ramsey, K.H.; Murthy, A.K. Peptide Vaccine: Progress and Challenges. Vaccines 2014, 2, 515–536. [Google Scholar] [CrossRef] [Green Version]
  202. Smith, P.L.; Piadel, K.; Dalgleish, A.G. Directing T-Cell Immune Responses for Cancer Vaccination and Immunotherapy. Vaccines 2021, 9, 1392. [Google Scholar] [CrossRef]
  203. Robbins, P.F.; Rosenberg, S.A.; Zhu, S.; Feldman, S.A.; Morgan, R.A. T cell Receptors Recognizing hla-a1-Restricted Mage-a3. U.S. Patent 2022/0195008 A1, 23 June 2022. [Google Scholar]
  204. Gey, A.; Tartour, E.; Bechard, D.U.S. Il-15 and il-15ralpha sushi Domain BASED Modulokines. U.S. Patent 2022/0193199 A1, 23 June 2022. [Google Scholar]
  205. Chen, Y.; Huang, X.; Kim, S. Combination Therapy for the Treatment of Cancer. U.S. Patent 2022/0193079 A1, 23 June 2022. [Google Scholar]
  206. June, C.H.; Zhao, Y.U.S. Rna Engineered t Cells for the Treatment of. Cancer. Patent 2022/0170012 A1, 22 June 2022. [Google Scholar]
  207. Roberts, D.D.; Pantoja, D.R.S. Methods for Modulating Chemotherapeutic Cytotoxicity. U.S. Patent 2022/0184111 A1, 16 June 2022. [Google Scholar]
  208. Korman, A.J.; Lonberg, N.; Fontana, D.J.; Gutierrez, A.A.; Selby, M.J.; Lewis, K. Combination of Anti-LAG-3 Antibodies and Anti-PD-1 Antibodies to Treat Tumors. U.S. Patent 2022/0185892 A1, 16 June 2022. [Google Scholar]
  209. Mortimer, S.A.W.; Talasaz, A.A.; Chudova, D.; Eltoukhy, H. Methods for Early Detection of Cancer. U.S. Patent 2022/0186323 A1, 16 June 2022. [Google Scholar]
  210. Clube, J. Selectively Altering Microbiota for Immune Modulation. U.S. Patent 11,291,723 A2, 5 April 2022. [Google Scholar]
  211. Mahr, A.; Weinschenk, T.; Schoor, O.; Fritsche, J.; Singh, H.; Song, C. Novel Peptides and Combination of Peptides and Scaffolds for Use in Immunotherapy Against Renal Cell Carcinoma (rcc) and Other Cancers. U.S. Patent 2022/0040278 A1, 10 February 2022. [Google Scholar]
  212. Manie, E.; Stern, M.-H.; Popova, T. Methods for Detecting Inactivation of the Homologous Recombination Pathway (brca1/2) in Human Tumors. U.S. Patent 2022/0010385 A1, 13 January 2022. [Google Scholar]
  213. Cobleigh, M.A.; Shak, S.; Baker, J.B. Gene Expression Markers for Breast Cancer Prognosis. U.S. Patent 11,220,715 B2, 11 January 2022. [Google Scholar]
  214. Fan, T.W.-M.; Lane, A.N.; Higashi, R.M.; Bousamra, M. Methods Related to Cancer. U.S. Patent 2022/0003792 A1, 6 January 2022. [Google Scholar]
  215. Diehn, M.; Alizadeh, A.A.; Newman, A.M. Identification and Use of Circulating Nucleic Acids. U.S. Patent 11,085,084 B2, 10 August 2021. [Google Scholar]
  216. Chen, J.; Buggy, J.J.; Elias, L. Methods for the Treatment of her2 Amplified Cancer. U.S. Patent 2021/0145835 A1, 20 May 2021. [Google Scholar]
  217. Kuntz, K.W.; Huang, K.-C.; Wook, H. Salt Form of a Human Histone Methyltransferase ezh2 Inhibitor. U.S. Patent 2021/0137936 A1, 13 May 2021. [Google Scholar]
  218. Vaske, C.J.; Benz, S.C.; Stuart, J.M. Pathway Recognition Algorithm Using Data Integration on Genomic Models (Paradigm). U.S. Patent 10,991,448 B2, 27 April 2021. [Google Scholar]
  219. Hoon, D.; Taback, B.; Shaolian, S. Method and Apparatus for in Vivo Surveillance of Circulating Biological Components. U.S. Patent 10,987,037 B2, 27 April 2021. [Google Scholar]
  220. Fyfe, G.; Phan, S.C.; Zhou, X. Anti-Angiogenesis Therapy for the Treatment of Breast Cancer. U.S. Patent 2021/0093715 A1, 1 April 2021. [Google Scholar]
  221. Paton, V.; Chirchir, A.B.; Klein, P. Treatment of Metastic Breast Cancer. U.S. Patent 2021/0047429 A1, 18 February 2021. [Google Scholar]
  222. Chui, S.; Smitt, M.; Patre, M. Methods of Treating her2-Positive Cancer. U.S. Patent 2021/0040216 A1, 11 February 2021. [Google Scholar]
  223. Knudsen, S. Methods for Predicting Drug Responsiveness in Cancer Patients. U.S. Patent 10,907,214 B2, 2 February 2021. [Google Scholar]
  224. Xiao, L.; Pu, C.; Cao, Z. Use of Chimeric Antigen Receptor Modified Cells to Treat Cancer. U.S. Patent 2020/0385484 A1, 10 December 2020. [Google Scholar]
  225. Doucey, M.-A.; Guex, N.; Crespo, I. Adoptive Immunotherapy for Treating Cancer. U.S. Patent 10,858,626b2, 8 December 2020. [Google Scholar]
  226. Pierce, D.; Carleton, M. Combination Therapy Comprising Nanoparticles of a Taxane and Albumin with abt-263 in Methods for Treating Cancer. U.S. Patent 2020/0246275 A1, 6 August 2020. [Google Scholar]
  227. Das-Young, L.; Wilner, K.D.; Nicholas, S. Combination of a PD-1 Antagonist and an ALK Inhibitor for Treating Cancer. U.S. Patent 10,695,426 B2, 30 June 2020. [Google Scholar]
  228. Mahr, A.; Weinschenk, T.; Schoor, O. Novel Peptides and Combination of Peptides for Use in Immunotherapy and Methods for Generating Scaffolds for the Use Against Pancreatic Cancer and Other Cancers. U.S. Patent 2020/0157177 A1, 21 May 2020. [Google Scholar]
  229. Adusumilli, P.S.; Sadelain, M.; Dimitrov, D.S. Mesothelin-Targeted Chimeric Antigen Receptors and Uses Thereof. U.S. Patent 10,633,441b2, 28 April 2020. [Google Scholar]
  230. Wang, D.; Jiang, W.; Agrawal, S. Treatment of Cancer Using tlr9 Agonist with Checkpoint Inhibitors. U.S. Patent 2020/0101102 A1, 2 April 2020. [Google Scholar]
  231. Markovic, S.N.; Nevala, W.K. Methods of Using Albumin-Antibody Nanoparticle Complex Compositions for Treating Cancer. U.S. Patent 10,596,112 B2, 24 March 2020. [Google Scholar]
  232. O’Donnell, J.; Bylesjo, M.; Patterson, F. Molecular Diagnostic Test for Cancer. U.S. Patent 10,378,066 B2, 13 August 2019. [Google Scholar]
  233. Themeli, M.; Sadelain, M.; Kloss, C.C. Effective Generation of Tumor-Targeted T Cells DERIVED from Pluripotent stem Cells. U.S. Patent 10,370,452 B2, 6 August 2019. [Google Scholar]
  234. Kipps, T.J.; Yu, J.; Cui, B. Antibodies and Vaccines for Use in Treating ROR1 Cancers and Inhibiting Metastasis. U.S. Patent 10,344,096 B2, 9 July 2019. [Google Scholar]
  235. Jensen, M.C. Drug Regulated Transgene Expression. U.S. Patent 10,266,592 B2, 23 April 2019. [Google Scholar]
  236. Mahr, A.; Weinschenk, T.; Hoerzer, H. Peptides and Combination of Peptides for Use in Immunotherapy Against BREAST Cancer and other Cancers. U.S. Patent 10,213,499 B2, 26 February 2019. [Google Scholar]
  237. Elsner, J.; Harris, R.L.; Lee, B.G. Substituted Pyrazino[2,3-b]pyrazines as mTOR Kinase Inhibitors. U.S. Patent 10,167,290 B2, 1 January 2019. [Google Scholar]
  238. Brentjens, R.J.; Jackson, H.J. Compositions and Methods for Immunotherapy. U.S. Patent 10,124,023 B2, 13 November 2018. [Google Scholar]
  239. Gurney, A.L.; Lewicki, J.; Satyal, S.H. Compositions and Methods for Diagnosing and Treating Cancer. U.S. Patent 2018/0222997 A1, 9 August 2018. [Google Scholar]
  240. Eltoukhy, H.; Talasaz, A.A. Methods and Systems for Detecting Genetic Variants. U.S. Patent 9,920,366 B2, 20 March 2018. [Google Scholar]
Figure 1. The process of immunoediting carried out by cancer cells.
Figure 1. The process of immunoediting carried out by cancer cells.
Diagnostics 13 02175 g001
Figure 2. Immune-suppression by tumour cells.
Figure 2. Immune-suppression by tumour cells.
Diagnostics 13 02175 g002
Figure 3. Mechanism of a peptide-based BCV.
Figure 3. Mechanism of a peptide-based BCV.
Diagnostics 13 02175 g003
Figure 4. Diagrammatic representation of vector-based vaccines.
Figure 4. Diagrammatic representation of vector-based vaccines.
Diagnostics 13 02175 g004
Table 1. Clinical trials in the field of breast cancer vaccines.
Table 1. Clinical trials in the field of breast cancer vaccines.
NCT NumberAntigens/BiologicalClinical Phase
NCT00854789E75 and GM-CSFI
NCT00892567Her-2/neu; CEA and CTAI
NCT02019524E39 and J65 peptidesI
NCT04270149ESR1 peptide vaccineI
NCT04521764Helicobacter pylori neutrophil-activating proteinI
NCT00343109HER-2/neuII
NCT02348320Personalised polyepitope DNA vaccineI
NCT02018458LA TNBC; ER+/HER-BCI/II
NCT04348747Anti-HER2/HER3 DC vaccine; PembrolizumabII
NCT02061423HER-2 pulsed DC vaccineI
NCT01730118AdHER-2/neu DC vaccineI
NCT00524277HER2-derived peptide GP2; GM-CSFII
NCT01479244HER2-derived peptide E75; GM-CSFI/II
NCT01570036HER2-derived peptide E75; GM-CSF; TrastuzumabII
NCT00140738HER; AS 15I/II
NCT02061332HER; DC vaccineII
NCT00399529HER2; GM-CSF; Cyclophosphamide; TrastuzumabII
NCT01479244HER2-derived peptide E75; GM-CSFIII
Table 2. Some patents related to cancer diagnosis and treatment filed/granted in the past five years.
Table 2. Some patents related to cancer diagnosis and treatment filed/granted in the past five years.
S. No.Patent NumberDate of PublicationInvention Disclosed
1.US 2022/0195008 A123 June 2022This invention disclosed the antigenic specificity of the T-cell receptor (TCR) for the melanoma antigen family. The polypeptide in the functional portion of the TCR was found to carry amino acid sequences of length 16–21 [203].
2.US 2022/0193199 A123 June 2022This invention described an immune cytokine that is a conjugate and an immunomodulatory antibody. It comprised an interleukin15-containing polypeptide, which is an IL-15Ra sushi domain-containing polypeptide. The immunomodulatory antibody- or antigen-binding fragment was said to be capable of binding PD-1 and PD-L1/L2 [204].
3.US 2022/0193079 A123 June 2022A pharmaceutical combination comprising a CDK inhibitor and an antihormonal agent that regulates the P13K/Akt/m TOR pathway or a pharmaceutically acceptable salt was discussed in the patent [205].
4.US 2022/0170012 A122 June 2022This invention disclosed the compositions and methods of generating an RNA chimeric-antigen receptor of transfected T-cells for use in adoptive therapy for cancer. The method modified the 5′ end of the RNA or its 7-methyl guanosine cap by the addition of the 5′-end of the eukaryotic messenger, soon after the initiation of the transcription [206].
5.US 2022/0184111 A116 June 2022Methods for reducing the cytotoxicity of chemotherapeutic agents towards non-cancer cells and increasing their cytotoxicity towards cancer cells were described in this patent. The patent gave details regarding the administration of an effective amount of the agent to achieve the inhibition of CD47 signalling for an effective chemotherapeutic agent [207].
6.US 2022/0185892 A116 June 2022This patent disclosed a method for the treatment of a solid tumour through the administration of an effective quantity of some anti-LAG-3 and anti-PD1 antibodies that carry the CD-R1, -R2, and -R3 domains of the chain [208].
7.US 2022/0186323 A1Jun16, 2022This invention discussed a method for detecting a tumour marker, which, in turn, indicated the presence of a cancer. The marker may be a sample of nucleic acid. The sample was then amplified by polymerase chain reaction, thereby enriching the nucleic acids for the detection of genomic regions [209].
8.US 11,291,723B25 April 2022A treatment method for enhancing the efficacy of a therapy for cancer in humans and animals was presented in this invention. This treatment was selective in killing or reducing the growth of the target cell by using the enzyme- Cas nuclease. It was also related to cell populations, system, arrays, cells, RNA, and other means affecting the therapy [210].
9.US 2022/0040278 A110 February 2022This patent disclosed cancer immunotherapy relating to tumour-associated T-cell peptide epitopes. The presence of peptides on the tested tissue sample biopsies helped to diagnose cancer. This patent also gave methods, such as antibody detection or spectrometry, for analysing peptides [211].
10.US 2022/0010385 A113 January 2022This invention discussed a method to detect the inactivation of the DNA homologous recombination pathway, which helped to detect the BC gene, BRCA, or placental alkaline phosphatase, PALP (also called FANCN), inactivation, which was probably due to a somatic mutation or a mutation in the germ cell line [212].
11.US 11,220,715B211 January 2022An expression of a set of genes as thera-prognostics for the disease-free survival of cancer patients was disclosed in this invention. It gives the use of a paraffin-embedded biopsy material that is compatible with different methods of tumour tissue harvest [213].
12.US 2022/0003792 A16 January 2022This invention mentioned the methods to determine the presence or absence of a cancer type in an animal. The procedure determined the concentration of lipid amounts in a sample, which was obtained either from a detected cancer or a treated individual’s body fluid, using the mass spectrometric technique [214].
13.US 11,085,084B210 August 2021This patent gave details regarding the identification and analysis of some polynucleotide adaptors, which include cell-free nucleic acids, from sample cancer patients. It also included their detection, diagnosis, and prognosis [215].
14.US 2021/0145835 A120 May 2021A pharmaceutical composition that could be utilised for the treatment of patients diagnosed with HER2-positive BC was addressed. The pharmaceutical substance disclosed was (R)-1-(3-(4-amino-3-(4-phenoxyphenyl)-1H pyrazolo [3,4-d] pyrimidin-1-yl) piperidin-1-yl) prop-2-en-1-one [216].
15.US 2021/0137936 A113 May 2021This invention described the administration of a therapeutically effective quantity of N-((4,6- dimethyl-2-oxo-1,2-dihydropyridin-3-yl) methyl) -5-(ethyl (tetrahydro-2H-pyran-4-yl) amino)-4- methyl-4′-(morpholino-methyl)-[1,1′-biphenyl]-3-carboxamide hydrobromide. The compound was a polymorph and exhibited an X-ray powder diffraction pattern with characteristic peaks [217].
16.US 10,991,448B227 April 2021This patent related to methods for the evaluation of the diagnosis of cancer in a patient. The patient may be subjected to some therapy. The attributes that were measured included the degree of mutation, transcription and translation levels, protein, and interaction. Thus, a probabilistic pathway gave a comparative graph representing the cellular processes [218].
17.US 10,987,037 B227 April 2021This invention was related to the collection of live tumour cells using a collecting probe, where the procedure involved placing the probe in a living organism. The probe used was composed of guide wire carrying a binding surface with an optically sensitive dye and an atraumatic tip at the distal end [219].
18.US 2021/0093715 A11 April 2021A method for the treatment of patients with metastatic BC that would extend the progression-free survival of the subjects under treatment was disclosed. The therapeutic regime consisted of an effective dose of a chemotherapeutic agent and an anti-VEGF antibody [220].
19.US 2021/0047429 A118 February 2021This invention disclosed a method for the treatment of BC with an effective amount of HER2 antibody and a taxane derivative [221].
20.US 2021/0040216 A111 February 2021This patent described a method for the treatment of patients diagnosed with HER2-positive BC. The method involved the administration of a therapeutic amount of an antagonist of programmed cell-death (PCD) protein-1 combined with Trastuzumab and Pertuzumab [222].
21.US 10,907,214B22 February 2021Certain methods, devices, and kits to be used for the detection of biomarkers in cancer patients were disclosed in this patent. The treatment involved a secretory phospholipase A and a hydrolysable cisplatin-containing liposome. It also discussed the use of a microarray device with an oligonucleotidic probe to assess the responsiveness of the patient [223].
22.US 2020/0385484 A110 December 2020The compositions and procedures for treating cancer with chimeric antigen receptor-modified cells were included in the invention. An antigen-binding domain, a transmembrane domain, a co-stimulatory signalling region, and a CD3 zeta signalling domain may all be present in the antigen [224].
23.US 10,858,626B28 December 2020This invention provided methods for use in the adoptive immunotherapy of diagnosed cancer patients. It gave the composition, which consisted of specific tumour infiltrating lymphocytes, tyrosine-protein kinase-2 receptor, and a vascular endothelial growth factor recepto inhibitor [225].
24.US 2020/0246275 A16 August 2020This invention gave the methods for the treatment of an individual with cancer, which comprised the administration of a therapeutically effective amount of a nanoformulation of some taxanes with albumin and a Bcl-2 inhibitor such as ABT-263 [226].
25.US 10,695,426B230 June 2020This invention described a combination therapy consisting of antagonists of PCD-1 and lymphoma kinase for use in the treatment of cancer. The treatment consisted of monoclonal antibody with variable regions of seq. ID nos. 13 and 15 with a heavy chain and a light chain [227].
26.US 2020/0157177 A121 May 2020An immunotherapeutic strategy for cancer was discussed in this invention. It described the use of tumour-associated T-cell peptide epitopes in combination with certain other associated peptide molecules bound to the major histocompatibility complex [228].
27.US 10,633,441B228 April 2020This invention claimed that methods and compositions enhance the immune system response towards cancers. It related antigen receptors that selectively target human mesothelin [229].
28.US 2020/0101102 A12 April 2020This patent gave methods for immune response induction in cancer patients using Toll-like receptor-9 agonists being administered intratumourally [230].
29.US 10, 596, 112B224 March 2020This invention disclosed the composition and methods of making antibodies and carrier proteins to be used as cancer therapeutic agents. It also discussed a lyophilisation procedure for nanoparticle formulation that contained albumin, antibodies, and some drug such as Paclitaxel [231].
30.US 10, 378, 066B213 August 2019A molecular diagnostic test for cancer was presented in this invention. The test was based on the detection of DNA damage and the repair mechanism and was capable of determining cancer in clinically responsive or non-responsive patients [232].
31.US 10, 370, 452B26 August 2019This invention related to immunotherapeutics obtained from pluripotent stem cells to generate some phenotypical, functionally expandable T-cells. The cells could be used to target a specific antigen, thereby enhancing the cytotoxic potential, antitumour activity, and, thus, the survival of the patient [233].
32.US 10, 344, 096B29 July 2019This patent disclosed some pharmaceutical compositions and their preparation methods. The composition was said to be useful in inhibiting metastasis using receptor tyrosine kinase-like orphan receptor1 antibodies (ROR1-antibodies) [234].
33.US 10, 266, 592B223 April 2019This invention described surface-engineered human T-lymphocytes for augmenting the immune response. The designed molecules helped mediate cellular immunotherapy and, thus, were an effective strategy to treat cancer [235].
34.US 10, 213, 499B226 February 2019The use of proteins, peptides, and nucleic acids such as immunotherapeutic agents, either alone or in combination, was presented. The peptides could either be peptide epitopes or bound to molecules such as the major histocompatibility complex [236].
35.US 10, 167, 290B21 January 2019This invention reported substituted pyrazino [2,3-b] pyrazines as inhibitors of mammalian target of rapamycin (mTOR) kinase. These compounds are used for the treatment of diseases such as cancer, inflammation, or neurogenerative disease [237].
36.US 10, 124, 023B213 November 2018This patent explored the use of certain cells such as immune-responsive cells, natural killer cells, T-lymphocytes, and regulatory T-cells, which were capable of expressing an antigen-binding receptor in the activation of immune cells. These were single-chain variable fragments that bound to the antigens with immunosuppressive activity, thereby reducing the suppressive action of the antigen [238].
37.US2018/0222997 A19 August 2018Certain novel compositions and methods to diagnose and treat solid tumours by the use of antibodies were described. It was said that the antibody was specifically bound to the extracellular domain of the human Frizzled (FZD) receptors, thereby inhibiting growth of tumour cells [239].
38.US 9, 920, 366B220 March 2018This invention described the methods and systems for determining genetic variants and detecting double-stranded DNA [240].
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

Fatima, G.N.; Fatma, H.; Saraf, S.K. Vaccines in Breast Cancer: Challenges and Breakthroughs. Diagnostics 2023, 13, 2175. https://doi.org/10.3390/diagnostics13132175

AMA Style

Fatima GN, Fatma H, Saraf SK. Vaccines in Breast Cancer: Challenges and Breakthroughs. Diagnostics. 2023; 13(13):2175. https://doi.org/10.3390/diagnostics13132175

Chicago/Turabian Style

Fatima, Gul Naz, Hera Fatma, and Shailendra K. Saraf. 2023. "Vaccines in Breast Cancer: Challenges and Breakthroughs" Diagnostics 13, no. 13: 2175. https://doi.org/10.3390/diagnostics13132175

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