Nothing Special   »   [go: up one dir, main page]

Next Article in Journal
The HuRaA Trial—The Radiocapitellar Line Shows Significant Posterior Translation in Healthy Elbows: A Prospective Analysis of 53 Healthy Individuals
Previous Article in Journal
The Role of microRNA-155 as a Biomarker in Diffuse Large B-Cell Lymphoma
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Diagnostics and Therapy for Malignant Tumors

1
Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
2
Department of Dentistry, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
3
Division of Nephrology, Department of Internal Medicine, Sijhih Cathay General Hospital, New Taipei City 221, Taiwan
4
School of Biological Sciences, University of California, San Diego, CA 92093, USA
5
Institute of Oral Medicine and Materials, College of Medicine, Tzu Chi University, Hualien 970, Taiwan
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomedicines 2024, 12(12), 2659; https://doi.org/10.3390/biomedicines12122659
Submission received: 31 October 2024 / Revised: 20 November 2024 / Accepted: 20 November 2024 / Published: 21 November 2024
(This article belongs to the Special Issue Diagnostics and Therapy for Malignant Tumors: New Developments)

Abstract

:
Malignant tumors remain one of the most significant global health challenges and contribute to high mortality rates across various cancer types. The complex nature of these tumors requires multifaceted diagnostic and therapeutic approaches. This review explores current advancements in diagnostic methods, including molecular imaging, biomarkers, and liquid biopsies. It also delves into the evolution of therapeutic strategies, including surgery, chemotherapy, radiation therapy, and novel targeted therapies such as immunotherapy and gene therapy. Although significant progress has been made in the understanding of cancer biology, the future of oncology lies in the integration of precision medicine, improved diagnostic tools, and personalized therapeutic approaches that address tumor heterogeneity. This review aims to provide a comprehensive overview of the current state of cancer diagnostics and treatments while highlighting emerging trends and challenges that lie ahead.

1. Introduction

Malignant tumors, which are characterized by uncontrolled cellular proliferation, invasion, and metastasis, are a major cause of morbidity and mortality worldwide [1]. Despite significant advancements in early detection and treatment of malignant tumors, the global burden of cancer continues to rise [2]. This can be attributed to factors such as the aging population, environmental changes, and lifestyle [3]. Modern oncology is advancing rapidly with the introduction of personalized medicine, immunotherapy, and novel diagnostic methods; however, challenges remain in ensuring equitable access to these technologies and translating scientific breakthroughs into clinical practice [4]. This review provides a detailed analysis of conventional and emerging diagnostic and therapeutic modalities in oncology.

2. Pathophysiology of Malignant Tumors

The development of malignant tumors, or carcinogenesis, is a multistep process involving genetic mutations, epigenetic alterations, and interactions between cancer cells and their microenvironment [5]. Key processes include apoptosis evasion, sustained angiogenesis, tissue invasion, and metastasis [6]. Genetic mutations in oncogenes (e.g., RAS and MYC) and tumor suppressor genes (e.g., TP53 and BRCA1) are often the driving factors [7]. Beyond these characteristics of malignant tumors mentioned earlier, immune evasion and metabolic reprogramming make cancer cells more aggressive within the human body. Cancer cells evade immune system attacks, including those from T cells and natural killer (NK) cells, which support their survival [8]. In colon cancer, a scarcity of killer lymphocytes is linked to poorer prognosis compared to patients with abundant T cells [9]. Additionally, the reprogramming of energy metabolism enables cancer cells to adapt to substantial alterations in the tumor microenvironment, which are driven by growth factors and hypoxia-induced factors [8]. Through the Warburg effect, cancer cells rely on fermentation to generate energy and sustain their high proliferation capacity [10]. Thus, these two characteristics, avoiding immune destruction and deregulating cellular energetics, can further facilitate tumor progression [11].

3. Diagnostic Techniques in Malignant Tumors

Accurate and early diagnosis is crucial for improving cancer outcomes [12]. Diagnostic approaches such as imaging techniques and molecular diagnostics have evolved significantly, incorporating both traditional and advanced techniques [13].

3.1. Imaging Techniques

Computed tomography (CT) and magnetic resonance imaging (MRI) remain as the cornerstone imaging techniques for tumor detection and staging. They offer high-resolution imaging, enabling detailed tumor characterization [14]. Paudyal et al. highlighted the increasing utility of artificial intelligence (AI) in enhancing tumor detection, segmentation, and monitoring, contributing to more precise and personalized cancer treatment. This review discusses how AI can streamline clinical workflows, improve image quality, and address the need for quantitative metrics beyond traditional lesion size measurements. However, it also identifies significant challenges, including data accuracy, bias, and the integration of AI tools into clinical practice in the near future [15]. Positron emission tomography (PET) scans, often combined with CT, are valuable for detecting metastasis by highlighting areas of high metabolic activity typical of cancer cells [16]. PET is particularly useful for monitoring responses to therapy [17]. Illimoottil and Ginat explored the application of deep learning (DL) to improve the diagnosis, treatment, and prognosis of head and neck cancers using imaging techniques such as MRI, CT, and PET. It highlights how advanced DL models, such as convolutional neural networks, generative adversarial networks, and transformer models, can enhance tumor detection, segmentation, and outcome prediction. These methods address some limitations of traditional imaging, including the subjectivity and variability in human interpretation [18]. Ultrasound and endoscopy have been reported to be useful medical technologies for certain cancers, such as pancreatic and gastrointestinal cancers, and they allow real-time imaging coupled with biopsy capabilities [19,20]. Qi et al. described the development and application of a novel endoscopic imaging technique called surgical polarimetric endoscopy (SPE). SPE uses polarized light to enhance the detection of laryngeal cancer, providing greater contrast between cancerous and healthy tissues than with traditional white light endoscopy. By measuring the differences in light polarization, SPE offers real-time, high-definition images that significantly improve cancer detection accuracy, particularly for early-stage and precancerous lesions that are difficult to distinguish using standard methods [21].
Therefore, CT and MRI remain essential for tumor detection and staging, providing high-resolution imaging that allows for detailed characterization of malignancies. The integration of AI and DL is transforming oncological imaging, enhancing tumor detection, segmentation, and monitoring while also addressing limitations such as variability in human interpretation. PET, particularly in combination with CT, plays a critical role in identifying metastases and monitoring therapeutic responses. Emerging techniques such as SPE offer promising advancements in early cancer detection by significantly improving the contrast between cancerous and healthy tissues. Despite this progress, challenges such as data accuracy, bias, and the integration of AI into clinical practice must be addressed to fully realize the potential of these technologies.

3.2. Molecular Diagnostics

Molecular diagnostics have become a cornerstone in the detection, classification, and management of cancer [22]. This approach uses genomic, transcriptomic, proteomic, and epigenetic alterations in cancer cells to provide precise and individualized insights into the biology of the disease [23,24]. Rapid advancements in molecular diagnostics have enabled earlier detection, more accurate prognosis, and the development of personalized treatment strategies [25]. This section covers the key molecular diagnostic tools currently used in oncology, including tumor biomarkers, tissue biopsies, and liquid biopsies.

3.2.1. Tumor Biomarkers for Diagnosis

Tumor biomarkers play a critical role in cancer diagnosis by providing measurable indicators of the presence or progression of malignancies [26]. These biomarkers are typically found in blood, urine, or tissues and are produced either by the tumor itself or by the body in response to cancer [27]. Identification and monitoring of these biomarkers have revolutionized cancer diagnostics by enabling early detection, improving prognosis, and guiding therapeutic decisions [28]. Although blood-detected biomarker levels need to be combined with other specific tests, including magnetic resonance imaging, transvaginal ultrasound, computed tomography, and additional biomarkers to achieve greater accuracy, they still hold credible diagnostic ability for specific cancers. Tumor biomarkers can be classified into several categories based on their molecular nature and diagnostic utility, including proteins, nucleic acids (genetics), lipids, metabolites, long non-coding RNAs (lncRNAs), and microRNAs (miRNAs) [27]. In this section, we summarize the various types of tumor biomarkers (Table 1, Table 2 and Table 3) and their applications in clinical oncology.
Table 1. Potential biomarkers for diagnosed cancer type.
Table 1. Potential biomarkers for diagnosed cancer type.
MoleculeBiomarkerDiagnostic ValueDiagnosed Cancer TypeReference
ProteinPSA2–10 ng/mLProstate cancer[29,30]
CA-125>30 U/mLOvarian cancer[31,32]
CEA5–10 μg/LColorectal cancer[33,34]
AFP>20 ng/mLHepatocellular carcinoma and germ cell tumor[35,36]
Nucleic acidBRCA1, BRCA218 ng/mLBreast cancer and Ovarian cancer[37,38]
EGFR197 copies/μLNon-small cell lung cancer[39,40]
KRAS>0.8 ng/μL (pancreatic cancer); 5450 alleles/mL (lung cancer)Colorectal cancer, pancreatic cancer, and lung cancer[41,42,43]
Methylated SEPT916 copies/mLColorectal cancer[44,45]
Methylated MGMT25.2 ng/mLGlioblastoma[46,47]
LipidLPA3.5 μmol/L (ovarian cancer); 0.1 μmol/L (breast cancer); 2.58 nmol/mL (prostate cancer)Ovarian cancer, breast cancer, and prostate cancer[47,48,49,50]
PC>0.28 μmol/LBreast cancer, liver cancer, and colorectal cancer[51,52,53,54]
S1P75–1100 nMBreast cancer, ovarian cancer, and colorectal cancer[55,56,57]
Ceramide0.00744 pmol/mgBreast cancer[58,59]
27-HC0.31 μMBreast cancer[60,61]
Cholesterol esterNo reportProstate cancer and glioblastoma[62,63]
FFA>0.4 mmol/LBreast cancer and prostate cancer[64,65,66]
Protein biomarkers are some of the most commonly used biomarkers in clinical practice. They are typically found in blood or tissues and can indicate the presence of cancer [67]. Prostate-specific antigen (PSA) is a well-known biomarker for the early detection of prostate cancer. Elevated PSA levels (2–10 ng/mL) in the blood can indicate the presence of prostate cancer, although benign conditions such as prostatitis or benign prostatic hyperplasia can also elevate PSA levels [29]. CA-125 is widely used to detect ovarian cancer. Elevated CA-125 levels (>30 U/mL) can indicate the presence of ovarian cancer; however, they are also elevated in other conditions, such as endometriosis, making it more useful for monitoring treatment response rather than for early detection [31]. Carcinoembryonic antigen (CEA) is primarily used as a biomarker for colorectal cancer, with levels ranging from 5 to 10 μg/L, but it can also be elevated in other cancers, such as breast, lung, and pancreatic cancers. It is typically used to monitor the disease progression and recurrence after treatment [33]. Alpha-fetoprotein (AFP) is a diagnostic marker for liver cancer (hepatocellular carcinoma) and germ cell tumors. AFP levels above 20 ng/mL in the blood can indicate the presence of these cancers, particularly in patients with underlying liver diseases [35].
Genetic tumor biomarkers are characterized by changes in DNA or RNA that indicate the presence of cancer. Genetic biomarkers provide insights into the molecular alterations driving tumor growth and can guide targeted therapies [68]. Mutations in the breast cancer gene 1 (BRCA1) and breast cancer gene 2 (BRCA2) genes significantly increase the risk of breast and ovarian cancers [69]. Testing for these mutations can guide preventive measures and inform treatment decisions, particularly for poly ADP-ribose polymerase inhibitors [37]. Epidermal growth factor receptor (EGFR) mutations are commonly found in non-small cell lung cancer [39]. Detecting these mutations can help select patients for targeted therapy with EGFR inhibitors such as erlotinib and gefitinib [41]. Kirsten rat sarcoma virus mutations are commonly observed in colorectal, pancreatic, and lung cancers [41]. Kirsten rat sarcoma virus mutations are often associated with resistance to certain targeted therapies such as anti-EGFR monoclonal antibodies in colorectal cancer [70]. Moreover, emerging biomarkers are used to assess genetic predispositions (BRCA mutations) and guide treatment options (human EGFR 2 in breast cancer and EGFR in lung cancer) [71]. Moreover, blood-based epigenetic biomarkers are used to reduce sampling invasiveness, simplify sampling procedures, and detect changes in gene expression without altering the DNA sequence itself, often through mechanisms such as DNA methylation or histone modification [72,73]. Methylated septin 9 DNA is a biomarker used in the detection of colorectal cancer through blood tests [44]. It is particularly useful for screening individuals at risk of colorectal cancer and offers a non-invasive alternative to colonoscopy [74]. Methylation of the O6-methylguanine-DNA methyltransferase gene promoter is a predictive biomarker of glioblastoma [46]. Tumors with this epigenetic alteration respond better to alkylating agents, such as temozolomide, because methylation silences the DNA repair gene, making the tumor more susceptible to DNA-damaging therapies [75]. These biomarkers will pave the way for more personalized and effective cancer treatments.
Lipids play critical roles in cancer biology, serving not only as structural components of cell membranes but also as signaling molecules that regulate cellular processes such as proliferation, apoptosis, and metabolism [76]. Aberrant lipid metabolism is a hallmark of cancer, and changes in the lipid composition and profile have been increasingly recognized as potential biomarkers for cancer diagnosis, prognosis, and therapeutic targeting [77]. Lipid-based tumor biomarkers offer promising insights into the metabolic alterations that occur in cancer cells and can serve as non-invasive diagnostic tools through blood or tissue analysis [76]. A discussion of the clinical applications of lipid biomarkers in cancer is presented in this section. Lipids are a diverse group of molecules. Different classes of lipids, including phospholipids, sphingolipids, fatty acids, and cholesterol derivatives, have been identified as potential biomarkers of various cancers [78]. Phospholipids, which constitute the structural backbone of cell membranes, show altered profiles in cancer cells due to changes in membrane synthesis and cellular signaling [79]. Lysophosphatidic acid (LPA) is a bioactive phospholipid that acts as a signaling molecule involved in cell growth, survival, and migration [80]. Increased levels of LPA have been detected in ovarian (3.5 μmol/L), breast (0.1 μmol/L), and prostate cancers (2.58 nmol/mL). It is particularly significant in ovarian cancer, where LPA levels in the plasma and ascitic fluid are often elevated, making it a potential biomarker for early detection [48,49]. Phosphatidylcholines (PCs) are the most abundant phospholipids in the cell membrane [81]. Alterations in PC metabolism, especially in levels of certain PC species, have been linked to breast, liver, and colorectal cancers. These alterations reflect the metabolic reprogramming of cancer cells to support rapid cell growth and membrane biogenesis [51,52,53]. Sphingolipids are involved in regulating cell death, proliferation, and differentiation. The dysregulation of sphingolipid metabolism is associated with cancer progression [82,83]. Sphingosine-1-phosphate (S1P) is a signaling sphingolipid that promotes tumor growth, angiogenesis, and metastasis by interacting with the S1P receptors on cancer cells. Elevated levels of S1P, ranging from 75 to 1100 nM, have been detected in breast, ovarian, and colorectal cancers, and are considered potential biomarkers of cancer progression and metastasis [55,56]. Ceramides, a subclass of sphingolipids, are involved in apoptotic signaling pathways [84]. During carcinogenesis, the balance between ceramides and S1P is often disrupted, which leads to increased cell survival [85]. Reduced ceramide levels are linked to chemotherapy resistance in certain cancers, such as breast cancer, making ceramide a biomarker of therapeutic response [58].
Cholesterol and its metabolites play essential roles in cancer cell membrane structure and signaling pathways [86]. Oxysterols are oxidized derivatives of cholesterol involved in the regulation of immune responses and cell proliferation [87]. Elevated levels of specific oxysterols, such as 27-hydroxycholesterol (27-HC), are associated with breast cancer progression, particularly in estrogen receptor-positive breast cancer, where they can mimic estrogen and promote tumor growth [60]. Increased levels of cholesterol esters have been found in aggressive forms of cancer, such as prostate cancer and glioblastoma [62,63]. Cholesterol ester accumulation reflects altered lipid metabolism in cancer cells driven by increased uptake and synthesis of cholesterol to support membrane production and oncogenic signaling [88]. Furthermore, fatty acids, both free and as components of complex lipids, are essential for cancer cell proliferation and survival [89]. Cancer cells often exhibit elevated levels of free fatty acids (FFAs) due to enhanced lipolysis and de novo fatty acid synthesis [90]. Elevated levels of certain FFAs above 0.4 mmol/L, such as palmitic and oleic acids, have been linked to breast and prostate cancers [64,65]. FFAs can be detected in blood samples and may serve as non-invasive biomarkers for cancer diagnosis and monitoring [91,92]. Alterations in the ratio of omega-6 to omega-3 polyunsaturated fatty acids (PUFAs) have been implicated in cancer progression [93]. Increased levels of omega-6 PUFAs, such as arachidonic acid, have been associated with inflammation and cancer growth, whereas omega-3 PUFAs, such as eicosapentaenoic acid, have been shown to have anti-inflammatory and anti-tumorigenic effects [94,95,96]. Taken together, the ratio of these fatty acids could serve as a potential biomarker of cancer risk and progression.
Metabolites are small molecules generated or consumed during metabolic processes. Metabolism encompasses both the breakdown of substances to produce energy (catabolism) and the synthesis of complex molecules (anabolism). Dysregulated cellular metabolism is a hallmark of cancer, leading to elevated levels of normal metabolites or the production of abnormal ones. These altered metabolite levels can serve as biomarkers to help predict cancer. Tumor cells can modify their metabolic pathways to support biosynthesis and meet energy demands, adapting to the influence of oncogenic mutations and changes in tumor suppressor functions [97,98]. To date, a significant body of evidence has shown that different types of metabolites exhibit abnormal levels in the blood (Table 2). However, identifying specific cancers based on circulating metabolite levels remains a major challenge in developing effective non-invasive diagnostic biomarkers.
Table 2. Metabolites as biomarkers for various cancer types.
Table 2. Metabolites as biomarkers for various cancer types.
Cancer TypeMetaboliteReference
Breast cancerCreatinine (↑), sarcosine (↑), 5-oxoproline (↑), L-phenylalanine (↑), glycoursodeoxycholic acid (↑), glycochenodeoxycholic acid, (↑) tauroursodeoxycholic acid (↑), 1-methylnicotimanide (↑), octanoic acid (↑), dodecanoylcarnitine (↑), L-acetylcarnitine (↑), docosahexaenoic acid (↑)[99]
Ovarian cancerarabitol (↑), maltose (↑), maltotriose (↑), raffinose (↑), mannitol (↑), demethylphylloquinone (↓), ganglioside (↑), N-formylkynurenine (↑), histidine (↓), citrulline (↓), citrate (↓), lysine (↑)[100]
Prostate cancerSarcosine (↑), kynurenine (↑), choline (↑), spermine (↑), citrate (↑), myo-inositol (↑), fructose (↑)[101,102]
Pancreatic cancercreatine (↑), inosine (↑), beta-sitosterol (↑), sphinganine (↑), glycocholic acid (↑), acetylcarnitine (↑), glutamine (↑), glutamic Acid (↓), symmetric dimethylarginine (↑), hexoses (↓)[103,104]
Gastric cancerGlucose (↓), lactate (↓), fumaric acid (↓), citrate (↑), α-ketoglutarate (↑), succinate (↓), pyruvic acid (↓), valine (↑), tryptophan (↓), leucine (↓), histidine (↓), glutamine (↑), gondoic acid (↑), palmitoleic acid (↑), cervonic acid (↑)[105]
Colon cancer5-hydroxytryptamine (↓), fumarate (↓), 4-hydroxystyrene (↓), hydroquinone (↓), cholic acid (↓), 2-hydroxy-3-methylpentanoic acid (↓), xanthosine (↑), sphinganine (↑), octenedioate (↑), β-hydroxybutyrate (↑), 2-oxobutanoic acid (↑)[106]
Bladder cancerIsobutyrate (↑), pyroglutamate (↑), propionate (↑), choline (↑), acetate (↑)[107]
Acute myeloid leukemia2-hydroxyglutarate (↑)[108]
Thyroid cancerCholine (↑), glucose (↑), mannose (↑), pyruvate (↑), 3-hydroxybutyric acid (↑), valine (↓), tyrosine (↓), proline (↓), lysine (↓), leucine (↓), gamma-aminobutyric acid (↑), aminooxyacetic acid (↑), 4-deoxypyridoxine (↑); pyroglutamic acid (↓)[109]
Liver cancerPutrescine (↑), cadaverine (↑), spermidine (↑), agmatine (↑), lysine (↑), arginine (↑), S-adenosyl-l-methionine (↑), N-acetylspermine (↑), N-acetylspermidine (↑), γ-aminobutyric acid (↑)[110]
Lung cancerPutrescine (↑), cadaverine (↑), spermidine (↑), agmatine (↑), ornithine (↑), lysine (↑), arginine (↑), S-adenosyl-l-methionine (↑), γ-aminobutyric acid (↑)[110]
↑: upregulation; ↓: downregulation.
In addition to these regular molecules as potential biomarkers for cancer diagnosis, lncRNAs and miRNAs have been found to exhibit differential expression between healthy individuals and cancer patients. lncRNAs are a type of RNA molecule defined as transcripts longer than 200 nucleotides that do not code for proteins but play crucial roles in regulating gene expression and cellular processes, including growth, differentiation, migration, and apoptosis. lncRNAs regulate the expression of oncogenes and tumor-suppressor genes in normal cells, and their dysregulation can promote tumorigenesis [111]. The levels of lncRNAs are associated with different types of cancer compared to normal tissue (Table 3). For instance, HOTAIR promotes metastasis in breast cancer by modifying chromatin states [112]. Conversely, the loss or downregulation of certain lncRNAs, such as MEG3, can disrupt critical cell growth regulation, contributing to cancer development [113]. Similar to lncRNAs, miRNAs, non-coding RNA molecules containing 21–23 nucleotides, have a role in cancer-associated biological processes [114]. As shown in Table 3, different miRNAs are associated with various types of cancer progression. Although the levels of both lncRNAs and miRNAs can change in the blood of cancer patients, a challenge remains in determining the specificity of these RNAs for diagnosing specific cancer types.
Table 3. lncRNA and miRNA as biomarkers for various cancer types.
Table 3. lncRNA and miRNA as biomarkers for various cancer types.
Cancer TypelncRNAmiRNAReference
Breast cancerZFAS1 (↓), LSINCT5 (↑), LINC00617 (↑), RP11-445H22.4 (↑), BC200 (↑), UCA1 (↑), SRA (↑), HOTAIR (↑)miR-126 (↓), miR-335 (↓), miR-199a (↓), Jet-7c (↓), Jet-7d (↓), miR-589 (↑), miR-425 (↑), miR-21 (↑), miR-34a (↑), miR-106a (↑), miR-195 (↑), Jet-7a (↑)[115,116,117,118,119,120,121,122,123,124,125]
GliomaTSLC1-AS1 (↓), ADAMTS9-AS2 (↓), MDC1-AS (↓), TUG1 (↓), ROR (↓), CACS2 (↓), GAS5 (↓), MEG3 (↓), XIST (↑), CRNDE (↑), MALAT1 (↑), HOTAIR (↑), HOXA11-AS (↑), Linc-POU3F3 (↑), ATB (↑), AB073614 (↑), H19 (↑), SPRY4-IT1 (↑)miR-29 (↓), miR-128 (↓), miR-205 (↓), miR-125b (↓), miR-122 (↓), miR-451a (↓), miR-203 (↓), miR-219-5p (↑), miR-21 (↑), miR-376c (↑), miR-210 (↑), miR-301a (↑), miR-454-3p (↑)[126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145]
Colorectal cancerLOC554202 (↓), PVT1 (↑), H19 (↑), AFAP1-AS1 (↑), MALAT1 (↑), CCAT1-L (↑), PCAT-1 (↑)miR-10b (↓), miR-155 (↓), miR-29a (↑), miR-92a (↑), miR-141 (↑), miR-221 (↑), Jet-7a (↑)[124,146,147,148,149,150,151,152,153,154,155]
Prostate cancerPTENP1 (↓), PCA3 (↑), PCAT5 (↑), PCAT18 (↑), PRNCR1 (↑), MALAT1 (↑), PCAT-1 (↑),miR-155 (↓), miR-21 (↑), miR-141 (↑), miR-221 (↑), miR-375 (↑), Jet-7a (↑)[124,156,157,158,159,160,161,162,163,164]
Gastric cancerLINC00152 (↑), LSINCT-5 (↑), H19 (↑), PVT1 (↑),Jet-7a (↓), miR-21 (↑), miR-106a (↑), miR-106b (↑), miR-17-5p (↑), miR-1 (↑), miR-34a (↑), miR-20 (↑), miR-27a (↑),[165,166,167,168,169,170]
Pancreatic cancerHULC (↑), HOTAIR (↑)miR-155 (↑), miR-21 (↑), miR-196a (↑), miR-210 (↑), miR-155 (↑), miR-200a (↑), miR-200b (↑),[171,172,173,174,175]
Lung cancerMALAT1 (↓), MEG3 (↓), UCA1 (↑), AFAP1-AS1 (↑), HOTAIR (↑), CCAT2 (↑), MVIH (↑), LCAL1 (↑), LUADT1 (↑)miR-30e-3p (↓), Jet-7f (↓), miR-1 (↓), miR-17-5p (↓), miR-27a (↓), miR-106a (↓), miR-146 (↓), miR-155 (↓), miR-221 (↓), miR-499 (↓), Jet-7a (↓), miR-21 (↑), miR-25 (↑), miR-29c (↑), miR-30d (↑), miR-223 (↑), miR-486 (↑)[113,119,176,177,178,179,180,181,182,183,184,185,186,187]
Hepatocellular cancerPRAL(↓), MALAT1 (↑), HOTAIR (↑), RP11-160H22.5 (↑), XLOC_014172 (↑), LOC149086 (↑), BANCR (↑), SNHG3 (↑), MVIH (↑), ANRIL (↑), HULC (↑)miR-92a (↓), miR-21 (↑), miR-122 (↑), miR-223 (↑), miR-500 (↑), miR-885-5p (↑)[188,189,190,191,192,193,194,195,196,197,198,199,200]
Oral cancerNEAT1 (↑), UCA1 (↑), HOTAIR (↑)miR-24 (↑), miR-31 (↑)[201,202,203,204,205]
Acute myeloid
leukemia
Wt1-as (↑)miR-92a (↓)[206,207]
Cervical cancerCCHE1 (↑), HOTAIR (↑), CCAT2 (↑)miR-1284 (↓), miR-573 (↓), miR-433 (↓), miR-424-5p (↓), miR-361-5p (↓), miR-383-5p (↓), miR-335-5p (↓), miR-874 (↓), miR-132 (↓), miR-411 (↓), miR-545 (↓), miR-143 (↓), miR-107 (↓), miR-1 (↓), miR-195 (↓), miR-31 (↑), miR-224 (↑), miR-92a (↑), miR-200a (↑), miR-96-5p (↑), miR-199b-5p (↑),[208,209,210,211]
MelanomaCASC15 (↑), SPRY4-IT1 (↑)miR-10b (↓), miR-155 (↓)[124,212,213,214]
Bladder cancerH19 (↑), UCA1 (↑)miR-21 (↑), miR-210 (↑), miR-29c (↓), miR-124 (↓), miR-29c (↓), miR-214 (↓), miR-29c (↓) [215,216,217]
↑: upregulation; ↓: downregulation.

3.2.2. Tissue and Liquid Biopsies for Tumor Diagnosis

Tissue biopsy is a critical component in the diagnosis and management of tumors. It involves the extraction of a tissue sample from a suspicious lesion or mass for histological examination. This process is essential to confirm the presence of cancer, determine its type, and guide treatment decisions. Biopsies provide critical information about a tumor’s histological and molecular characteristics, enabling oncologists to make informed decisions regarding treatment and management [218].
Liquid biopsies that analyze biomarkers from blood and other body fluids to detect cancer non-invasively are growing techniques for transforming cancer diagnosis and management [219]. Although traditional methods, such as tissue biopsies, remain the gold standard, liquid biopsies offer significant advantages, including being less invasive, more easily repeatable, and potentially more cost-effective. However, the current evidence on liquid biopsy technologies often lacks the sensitivity required to detect early-stage cancers [220]. Additionally, liquid biopsy analysis involves the identification of biomarkers for early cancer diagnosis, prognosis, therapeutic prediction, and follow-up by isolating circulating tumor cells, circulating tumor DNA, extracellular vesicles, and tumor-educated platelets from body fluid samples and analyzing their molecular characteristics [221,222].

3.2.3. Integration of Histopathology, Genomics, and Big Data for Personalized Cancer Diagnosis

Histopathology has been a crucial tool for cancer diagnosis and prognosis for over a century. It reveals key features for assessing cancer progression, including molecular insights and characteristics such as nuclear atypia, cell type, disease development, mitotic activity, genetic alterations, cell viability, and tissue architecture [223,224]. Integrating cytological details and complex tissue patterns helps to classify and grade lesions. Histopathology is essential for a precise diagnosis by integrating genomic information [225]. The identification of specific genetic events in tumors has been driven by the rapid growth of molecular diagnostics such as polymerase chain reactions and immunohistochemistry [225].
Molecular diagnostics are increasingly required to assist in diagnosing various tumors, transitioning from the study of tumor pathogenesis to applications in clinical laboratories [225]. To date, several tumor-related genes have been shown to be involved in cancer development (Table 4). For example, the identification of BRCA1 and BRCA2 mutations in breast cancer, mutL homolog 1, mutS homolog 6, and mutS homolog 6 mutations in colon cancer, and the RB1 mutation in retinoblastoma is useful for understanding cancer progression in relation to different prognoses and treatments [226,227,228]. However, point-by-point identification of tumor-related genes is not applicable to all patients, and the cost of personalized molecular identification is high. Thus, the integration of molecular profiling information with big data for personalized diagnosis is currently ongoing. To achieve this objective, the following tasks must be completed.
Table 4. Cancer types are caused by inherited mutant genes.
Table 4. Cancer types are caused by inherited mutant genes.
Cancer TypeInherited GenesReference
Breast cancerBRCA1, BRCA2, tumor protein P53 (TP53), phosphatase and tensin homolog (PTEN), mutY DNA glycosylase (MUTYH), serine/threonine kinase 11 (STK11)[229,230,231,232,233]
Ovarian cancerBRCA1, BRCA2, human mutL homolog 1 (MLH1), mutS homolog 2 (MSH2), mutS homolog 6 (MSH6), postmeiotic segregation increased 2 (PMS2), MUTYH, STK11, PTEN[229,231,232,233,234]
Prostate cancerBRCA2, MLH1, MSH2, MSH6, PMS2[235,236]
Pancreatic cancerBRCA1, BRCA2, adenomatous polyposis coli (APC), STK11, multiple endocrine neoplasia type 1 (MEN1), cyclin-dependent kinase inhibitor 2A (CDKN2A)[237,238,239]
Gastric cancerBRCA1, BRCA2, MLH1, MSH2, MSH6, PMS2, APC, STK11, MEN1[240,241,242,243,244]
Colon cancerMLH1, MSH2, MSH6, PMS2, PTEN, TP53, STK11[230,231,233,245]
Bladder cancerMLH1, MSH2, MSH6, PMS2, MUTYH[232,246]
Gallbladder cancerMLH1, MSH2, MSH6, PMS2, TP53[247]
Womb cancerMLH1, MSH2, MSH6, PMS2, PTEN, MUTYH, TP53[248,249,250]
GlioblastomaTP53, STK11[230,233]
Bone cancerTP53[251]
Acute myeloid leukemiaTP53[252]
Soft tissue sarcomaTP53, STK11[251]
MelanomaPTEN, STK11[233,253]
Thyroid cancerPTEN[231]
Kidney cancerPTEN[231]
Liver cancerAPC[254]
Retinoblastomaretinoblastoma 1 (RB1)[255]
Lung cancerTP53, STK11[230,233]
Esophageal cancerTP53[230]
Data Collection and Standardization: Histopathological and genomic data must be collected with high-resolution images, digitized, and then stored alongside DNA sequencing data, including mutations and gene expression profiles [223]. Histopathological images are analyzed using image recognition software, and machine learning to identify patterns and disease markers [223]. Finally, both data types must be standardized for integration.
Data Integration: Patient-specific histopathology images and genomic profiles are integrated into a unified framework for comprehensive analysis. This allows us to examine how genetic alterations manifest as tissue-level abnormalities. Machine learning models, including DL networks, can analyze image and sequence data to identify patterns and predict outcomes [256].
Machine Learning and AI Analysis: AI algorithms can detect cancer types, stages, and genetic mutations from histopathological images, while genomic data can predict outcomes based on mutations and gene expression [257]. Big data improve the accuracy of predictive models, enabling AI to correlate histopathological features with genetic mutations to forecast patient prognosis and treatment response.
Personalized Diagnosis and Treatment: Integration of genomic and histopathological data enables clinicians to identify cancer patterns and predict effective treatments for individual patients. Big-data-powered clinical decision support systems provide real-time diagnosis and treatment recommendations based on similar historical cases [258].
Research and Drug Development: Integrating big data from histopathology and genomics identifies novel biomarkers for early detection and targeted therapy while improving patient selection for clinical trials based on specific genetic profiles or histopathological features [259].
Data Sharing and Collaboration: Uploading anonymized data to global databases accelerates disease discovery and treatment by allowing researchers and healthcare providers to share resources. This collaboration pools data across regions, creating larger datasets that improve the model’s accuracy and robustness [260].
Real-Time Analytics: Real-time analytic tools enable clinicians to process big data quickly, allowing for faster and more accurate assessments of histopathological and genomic data [261]. Applying big data techniques allows the integration of histopathology and genomics into personalized healthcare systems, resulting in more accurate and data-driven diagnoses and treatments [262].
The integration of AI and big data in clinical oncology has transformative potential but is also accompanied by challenges and considerations specific to healthcare environments. AI-driven technologies, particularly machine learning (ML) and deep learning (DL), are increasingly applied in diagnostics and treatment planning [263]. These applications range from enhancing imaging interpretation, such as automated tumor detection and segmentation in MRI or CT scans, to real-time analysis of large-scale genomic data that aids in identifying molecular targets for personalized therapies [15]. Big data can combine patient demographics, imaging, molecular profiles, and outcomes, facilitating advanced predictive analytics that enhance treatment personalization and potentially improve prognostic accuracy [264]. Importantly, the challenges in clinical integration have been reported as associated with data accuracy/reliability, bias/generalizability, infrastructure requirements, and regulatory/ethical considerations. In data accuracy and reliability, clinical settings require high standards of accuracy, as errors in AI-driven diagnostics or treatment recommendations could lead to adverse patient outcomes [265]. AI models depend on high-quality, labeled datasets to achieve these standards, yet variability in data sources, imaging quality, and incomplete patient records can undermine model reliability [266]. In bias and generalizability, AI models trained on biased datasets (e.g., from specific populations) may not perform as well across diverse demographic groups, which could lead to disparities in care quality [267]. Ensuring AI systems are validated on diverse datasets is essential for equitable clinical application [268]. In infrastructure requirements, implementing AI in routine practice necessitates significant infrastructural investment, including data storage solutions for high volumes of imaging and genomic data, computational resources to handle complex analyses, and secure systems to protect patient privacy [269]. Additionally, healthcare providers need training to interpret AI outputs and integrate them into decision making [270]. In regulatory and ethical considerations, regulatory approval for AI models in healthcare remains a lengthy process, often requiring extensive validation studies [271]. Moreover, ethical guidelines for AI usage are still evolving, particularly regarding patient data usage and transparency in AI decision making [272]. Taken together, while AI and big data hold promise for enhancing oncology care, addressing these specific challenges is crucial to their safe, effective, and equitable integration into clinical practice.

4. Therapeutic Strategies for Malignant Tumors in Personalized and Targeted Therapies

In recent years, the treatment of malignant tumors has evolved drastically owing to advancements in personalized and targeted therapies [273]. These therapeutic strategies focus on tailoring treatments to the specific genetic, molecular, and environmental factors of each patient’s tumor, thereby increasing treatment efficacy and minimizing side effects. Personalized and targeted therapies have revolutionized cancer care, particularly for cancers that are resistant to traditional treatments such as chemotherapy and radiation [274,275]. This section outlines key approaches and advancements in personalized and targeted therapies for malignant tumors. The following sections explore the current and emerging treatment options.

4.1. Precision Molecular Oncology in Personalized Medicine

Precision molecular oncology is at the forefront of personalized medicine and is transforming approaches to cancer diagnosis, treatment, and management [276]. By analyzing the unique molecular characteristics of each patient’s tumor, including genetic, epigenetic, and proteomic alterations, precision oncology enables the development of highly individualized therapeutic strategies tailored to target specific drivers of cancer in each patient [277]. This approach contrasts with traditional “one-size-fits-all” treatments, focusing instead on the underlying biology of each tumor [278]. Next-generation sequencing (NGS) has revolutionized genomics, significantly advancing clinical diagnosis and precision medicine by allowing the rapid, cost-effective analysis of large-scale genetic data. NGS is a powerful tool for identifying disease-causing variants, enabling early and accurate diagnosis of genetic disorders, and facilitating the discovery of novel genes for targeted therapies. The combination of NGS and other omics, AI-driven analysis, would improve genome exploration and analysis for non-invasive monitoring in precision medicine [279,280,281]. Molecular imaging is a non-invasive medical technique that allows for the visualization, characterization, and quantification of biological processes at the molecular and cellular levels within tumors. In the field of theragnostic, molecular imaging offers a vision of how it may be combined with other diagnostic techniques to make cancer treatment more efficient and effective one day [282,283].

4.2. Surgery

Surgical resection is often the first-line treatment for localized solid tumors [284]. Advances in minimally invasive surgeries, such as robot-assisted procedures, have improved precision, reduced recovery times, and minimized complications [285]. For some cancers, such as breast cancer, sentinel lymph node biopsies have replaced full lymph node dissection, reducing morbidity [286].

4.3. Chemotherapy

Chemotherapy is a cancer treatment that uses cytotoxic drugs to target and kill rapidly dividing cancer cells by interfering with the synthesis of DNA, RNA, or proteins [287]. However, because chemotherapy lacks specificity, it also affects healthy cell growth, leading to side effects [288]. As a result, patients undergoing chemotherapy often experience complications such as anemia, caused by bone marrow suppression, and non-specific toxicities, including fatigue, hair loss, and mouth sores, due to damage to normal rapidly dividing tissues [287].
Several strategies can help minimize these complications. To address anemia, patients may use erythropoiesis-stimulating agents, such as epoetin, darbepoetin, and methoxy polyethylene glycol-epoetin β, which stimulate the bone marrow to produce more red blood cells [289]. For severe anemia, blood transfusions offer an effective way to increase red blood cell counts. If anemia is related to iron deficiency, iron supplements can also be beneficial [290].
For non-specific toxicity, drugs like amifostine can protect certain tissues from the toxic effects of chemotherapy, or symptom-relief medications may be used [291]. Patients can discuss with their doctor the option of optimizing dosage and frequency to balance efficacy and minimize toxicity. If alternative treatments are available, targeted therapy or immunotherapy can specifically target cancer cells, sparing healthy cells and reducing toxicity [292].
Although chemotherapy displays many side effects, combination chemotherapy remains a cornerstone treatment for many cancers, including hematological malignancies (e.g., acute myeloid leukemia) and solid tumors (e.g., breast, lung, and colorectal cancers) [293].

4.4. Radiation Therapy

Radiation therapy uses ionizing radiation to kill cancer cells by damaging DNA. Techniques such as intensity-modulated radiation therapy and stereotactic body radiotherapy have increased the precision of radiation delivery, minimizing damage to the surrounding healthy tissues. Proton therapy is an advanced form of radiation that offers even greater precision, particularly for tumors near critical structures (e.g., brain tumors) [294].

4.5. Targeted Therapy

Targeted therapies focus on the specific molecular targets that drive cancer growth. These treatments have shown significant success in cancers with known genetic mutations, including tyrosine kinase inhibitors and monoclonal antibodies [295]. For example, drugs such as imatinib for chronic myeloid leukemia and erlotinib for EGFR-mutated lung cancer have transformed treatment paradigms by specifically targeting cancer-driving mutations. Furthermore, antibodies such as trastuzumab (Herceptin) target HER2 in breast cancer, blocking the signaling pathways that promote cell proliferation [296].

4.6. Immunotherapy

Cancer therapy has undergone a paradigm shift with the advent of immunotherapy, a treatment modality that leverages the immune system to combat cancer [297] and is summarized in Figure 1. Unlike traditional therapies such as chemotherapy and radiation, which target tumor cells directly, immunotherapy enhances the body’s natural defense mechanisms [292]. The key players in cancer immunology include immune checkpoints, cytokines, and immune cells, such as T cells and natural killer (NK) cells [298]. Immune checkpoints, such as programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), play critical roles in maintaining immune homeostasis but are exploited by tumors to evade immune responses. Immune checkpoint inhibitors (ICIs), such as pembrolizumab (anti-PD-1) and ipilimumab (anti-CTLA-4), block these pathways, restoring T cell activity against tumor cells [299,300]. In clinical settings, ICIs have transformed the treatment landscape for cancers like melanoma, non-small cell lung cancer, and renal cell carcinoma. However, their efficacy is limited to patients with a pre-existing anti-tumor immune response [301]. The challenge with ICIs involves resistance mechanisms, such as mutations in antigen presentation machinery, and remain as significant barriers. Ongoing research focuses on combining ICIs with other therapies to overcome resistance [302].
There is potential for novel treatments for cancer, autoimmune diseases, and infectious diseases based on cytokine-based therapies [303]. Cytokines are crucial modulators of immune responses. Interleukin-2 (IL-2) and interferon-alpha (IFN-α) are among the first cytokines approved for cancer treatment [304]. It has been reported that high-dose IL-2 therapy promotes the expansion of cytotoxic T cells, leading to positive responses in metastatic melanoma and renal cell carcinoma [305]. On one hand, the advancements of cytokine-based therapy are to develop novel engineered cytokines aimed to enhance specificity and reduce systemic toxicity [306]. For example, IL-15 superagonists are being explored for their ability to activate NK and memory T cells [307]. On the other hand, adoptive cell therapies (ACTs) involve the genetic modification or expansion of immune cells to enhance their anti-tumor activity. Chimeric antigen receptor (CAR) T-cell therapy is the most notable example [308]. CAR T cells are engineered to recognize specific tumor antigens, such as CD19 in B-cell malignancies. Once infused, they proliferate and kill cancer cells [309]. However, challenges include cytokine release syndrome (CRS), neurotoxicity, and limited efficacy in solid tumors. Strategies to improve CAR T-cell trafficking and persistence are under investigation [310].
Cancer vaccines aim to elicit an immune response against tumor-specific or tumor-associated antigens. Examples include dendritic cell-based vaccines, such as sipuleucel-T (Provenge®) for prostate cancer [311]. The advantage of cancer vaccine is well known, neoantigen vaccines, personalized based on tumor mutational profiles, have shown promise in early-phase clinical trials [312]. In the near future, combining vaccines with checkpoint inhibitors may synergize immune activation [313]. Interestingly, oncolytic viruses selectively infect and kill tumor cells while stimulating anti-tumor immunity [314]. For example, Talimogene laherparepvec (T-VEC), a herpes simplex virus, has been approved for melanoma [315]. These viruses lyse tumor cells and release tumor antigens, enhancing immune system recognition [316]. Patients with cancer will be able to fully benefit from these emerging agents, which was made possible with T-VEC and other promising oncolytic viruses [317]. Taken together, immunology has revolutionized cancer therapy, providing durable responses in cases where traditional therapies fall short. Thus, continued research is needed to address challenges such as resistance, toxicity, and limited efficacy in certain cancers. The integration of immunotherapies with emerging technologies, such as artificial intelligence and genomics, promises to further enhance their potential.

4.7. Hormonal Therapy

Hormonal therapy is primarily used for cancers driven by hormones such as breast and prostate cancers. Drugs, such as tamoxifen (used for breast cancer) and androgen deprivation therapy (used for prostate cancer), block the effects of estrogen and testosterone, respectively, to slow tumor growth [318].

4.8. Gene Therapy

Gene therapy is an innovative approach to cancer treatment that involves modifying genetic material to treat or prevent the disease [319]. This strategy targets the underlying genetic and molecular abnormalities that drive cancer progression [320]. Unlike traditional therapies, gene therapy offers the potential for highly specific and long-lasting effects by directly altering the genetic makeup of cancer cells or enhancing the body’s natural defenses against tumors [321]. Many cancers arise from the loss or mutation of tumor suppressor genes, such as TP53 or RB1 [322]. Gene replacement therapy involves delivering functional copies of these genes to cancer cells using vectors, such as adenoviruses or lentiviruses, to restore normal cellular function [323]. Moreover, cancer cells are genetically modified to express a “suicide gene” such as the herpes simplex virus thymidine kinase (HSV-tk) [324]. When exposed to a prodrug like ganciclovir, these cells convert it into a toxic compound, selectively killing the tumor while sparing normal cells [325]. CRISPR-Cas9 technology enables precise editing of cancer-associated genes. It can be used to knockout oncogenes, repair tumor suppressor genes, or introduce genes that enhance immune cell recognition of tumors [326]. Therefore, gene therapy represents a transformative avenue in cancer treatment, offering precision and durability unmatched by conventional therapies. As delivery technologies improve and safety concerns are addressed, gene therapy holds immense potential to redefine the landscape of cancer treatment [327].
Despite significant advancements, cancer treatment still faces critical challenges, including tumor heterogeneity, drug resistance, and adverse effects [328]. Addressing these issues requires innovative strategies that account for tumor complexity, cancer cell adaptability, and patient well-being. Here are some targeted solutions:
  • Tumor Heterogeneity: cancers often consist of multiple subclones with different genetic profiles, leading to resistance to therapy and disease relapse;
    (1)
    Single-Cell Sequencing and Multi-Omics: applying single-cell sequencing, multi-omics profiling, and spatial transcriptomics enables the identification of diverse cancer cell populations within a tumor, guiding more precise treatment strategies [329];
    (2)
    Combination Therapy: simultaneously targeting multiple pathways through combination therapies can inhibit the survival of different subclones, reducing the likelihood of resistance [330];
    (3)
    Adaptive Treatment Approaches: adjusting treatment protocols in response to tumor evolution, known as adaptive therapy, helps control dominant subclones and delays resistance [331].
  • Drug Resistance: resistance to targeted therapies remains a major hurdle, as tumors can adapt by acquiring new mutations, activating alternative pathways, or altering drug targets [332];
    (1)
    Targeted Therapy Rotation: rotating between targeted therapies before resistance arises may lower the chances of cancer cells adapting to any single treatment [333];
    (2)
    Next-Generation Inhibitors: developing inhibitors targeting mutations linked to resistance mechanisms can prolong therapy effectiveness [334];
    (3)
    Targeting Alternative Pathways: drugs designed to block compensatory pathways that tumors activate can provide additional treatment options and prevent adaptation [335].
  • Adverse Effects: Many cancer treatments, including chemotherapy, radiation, and immunotherapy, carry significant side effects, impacting the quality life of patients. Balancing treatment efficacy with safety is a core concern [336].
    (1)
    Precision Medicine and Biomarkers: tailoring treatments based on biomarkers and genetic profiles allows for more targeted therapy, reducing side effects by matching treatments to individual tumor characteristics [275];
    (2)
    Prophylactic and Symptom-Management Strategies: integrating supportive care measures to manage side effects, such as antiemetics for chemotherapy-related nausea, improving the quality of life of patients [337];
    (3)
    Innovative Delivery Systems: utilizing advanced drug delivery systems, like nanoparticle carriers, that selectively target cancer cells can limit harm to healthy cells, reducing adverse effects [338].
These approaches offer a path forward in overcoming the persistent challenges in cancer treatment, aiming to improve both patient outcomes and quality of life.

5. Future Directions

The future of oncology lies in the integration of precision medicine, which tailors treatment based on the unique genetic makeup of both the patient and the tumor. Advances in bioinformatics, AI, and machine learning are likely to enhance the ability to analyze complex data, leading to more accurate predictions of treatment responses. Liquid biopsies and real-time monitoring of circulating tumor DNAs are likely to play increasingly important roles in tracking disease progression and treatment efficacy.

6. Conclusions

The landscape of cancer diagnostics and therapy has undergone tremendous advancements in recent decades. From traditional surgery and chemotherapy to novel approaches, such as immunotherapy and gene editing, the fight against cancer is more hopeful than ever. However, challenges such as tumor heterogeneity, drug resistance, and accessibility to advanced treatments persist. The integration of personalized medicine combined with ongoing research in cancer biology and innovative technologies promises to transform cancer care and offer better outcomes for patients globally (Figure 2).

Author Contributions

C.-C.T., C.-Y.W., H.-H.C., P.-C.H. and C.-Y.K.; Writing—original draft preparation and formal analysis: P.T.S.C., C.-H.C., T.Y.C., P.-C.H. and C.-Y.K.; Writing, Reviewing, and Editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, grant number TCRD-TPE-110-29.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Brown, J.S.; Amend, S.R.; Austin, R.H.; Gatenby, R.A.; Hammarlund, E.U.; Pienta, K.J. Updating the Definition of Cancer. Mol. Cancer Res. 2023, 21, 1142–1147. [Google Scholar] [CrossRef] [PubMed]
  2. Biemar, F.; Foti, M. Global progress against cancer-challenges and opportunities. Cancer Biol. Med. 2013, 10, 183–186. [Google Scholar] [CrossRef] [PubMed]
  3. Soerjomataram, I.; Shield, K.; Marant-Micallef, C.; Vignat, J.; Hill, C.; Rogel, A.; Menvielle, G.; Dossus, L.; Ormsby, J.N.; Rehm, J.; et al. Cancers related to lifestyle and environmental factors in France in 2015. Eur J Cancer 2018, 105, 103–113. [Google Scholar] [CrossRef] [PubMed]
  4. Masucci, M.; Karlsson, C.; Blomqvist, L.; Ernberg, I. Bridging the Divide: A Review on the Implementation of Personalized Cancer Medicine. J. Pers. Med. 2024, 14, 561. [Google Scholar] [CrossRef]
  5. Zhang, S.; Xiao, X.; Yi, Y.; Wang, X.; Zhu, L.; Shen, Y.; Lin, D.; Wu, C. Tumor initiation and early tumorigenesis: Molecular mechanisms and interventional targets. Signal Transduct. Target. Ther. 2024, 9, 149. [Google Scholar] [CrossRef]
  6. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef]
  7. Lee, E.Y.; Muller, W.J. Oncogenes and tumor suppressor genes. Cold Spring Harb. Perspect. Biol. 2010, 2, a003236. [Google Scholar] [CrossRef]
  8. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  9. Pagès, F.; Galon, J.; Dieu-Nosjean, M.C.; Tartour, E.; Sautès-Fridman, C.; Fridman, W.H. Immune infiltration in human tumors: A prognostic factor that should not be ignored. Oncogene 2010, 29, 1093–1102. [Google Scholar] [CrossRef]
  10. Navarro, C.; Ortega, Á.; Santeliz, R.; Garrido, B.; Chacín, M.; Galban, N.; Vera, I.; De Sanctis, J.B.; Bermúdez, V. Metabolic Reprogramming in Cancer Cells: Emerging Molecular Mechanisms and Novel Therapeutic Approaches. Pharmaceutics 2022, 14, 1303. [Google Scholar] [CrossRef]
  11. de Visser, K.E.; Joyce, J.A. The evolving tumor microenvironment: From cancer initiation to metastatic outgrowth. Cancer Cell 2023, 41, 374–403. [Google Scholar] [CrossRef] [PubMed]
  12. Crosby, D.; Bhatia, S.; Brindle, K.M.; Coussens, L.M.; Dive, C.; Emberton, M.; Esener, S.; Fitzgerald, R.C.; Gambhir, S.S.; Kuhn, P.; et al. Early detection of cancer. Science 2022, 375, eaay9040. [Google Scholar] [CrossRef] [PubMed]
  13. Pulumati, A.; Pulumati, A.; Dwarakanath, B.S.; Verma, A.; Papineni, R.V.L. Technological advancements in cancer diagnostics: Improvements and limitations. Cancer Rep. 2023, 6, e1764. [Google Scholar] [CrossRef] [PubMed]
  14. Rowe, S.P.; Pomper, M.G. Molecular imaging in oncology: Current impact and future directions. CA Cancer J. Clin. 2022, 72, 333–352. [Google Scholar] [CrossRef]
  15. Paudyal, R.; Shah, A.D.; Akin, O.; Do, R.K.G.; Konar, A.S.; Hatzoglou, V.; Mahmood, U.; Lee, N.; Wong, R.J.; Banerjee, S.; et al. Artificial Intelligence in CT and MR Imaging for Oncological Applications. Cancers 2023, 15, 2573. [Google Scholar] [CrossRef]
  16. Fiorentino, A.; Laudicella, R.; Ciurlia, E.; Annunziata, S.; Lancellotta, V.; Mapelli, P.; Tuscano, C.; Caobelli, F.; Evangelista, L.; Marino, L.; et al. Positron emission tomography with computed tomography imaging (PET/CT) for the radiotherapy planning definition of the biological target volume: PART 2. Crit. Rev. Oncol. Hematol. 2019, 139, 117–124. [Google Scholar] [CrossRef]
  17. Hicks, R.J. The role of PET in monitoring therapy. Cancer Imaging 2005, 5, 51–57. [Google Scholar] [CrossRef]
  18. Illimoottil, M.; Ginat, D. Recent Advances in Deep Learning and Medical Imaging for Head and Neck Cancer Treatment: MRI, CT, and PET Scans. Cancers 2023, 15, 3267. [Google Scholar] [CrossRef]
  19. Spadaccini, M.; Koleth, G.; Emmanuel, J.; Khalaf, K.; Facciorusso, A.; Grizzi, F.; Hassan, C.; Colombo, M.; Mangiavillano, B.; Fugazza, A.; et al. Enhanced endoscopic ultrasound imaging for pancreatic lesions: The road to artificial intelligence. World J. Gastroenterol. 2022, 28, 3814–3824. [Google Scholar] [CrossRef]
  20. Impellizzeri, G.; Donato, G.; De Angelis, C.; Pagano, N. Diagnostic Endoscopic Ultrasound (EUS) of the Luminal Gastrointestinal Tract. Diagnostics 2024, 14, 996. [Google Scholar] [CrossRef]
  21. Qi, J.; Tatla, T.; Nissanka-Jayasuriya, E.; Yuan, A.Y.; Stoyanov, D.; Elson, D.S. Surgical polarimetric endoscopy for the detection of laryngeal cancer. Nat. Biomed. Eng. 2023, 7, 971–985. [Google Scholar] [CrossRef] [PubMed]
  22. Riedl, J.M.; Moik, F.; Esterl, T.; Kostmann, S.M.; Gerger, A.; Jost, P.J. Molecular diagnostics tailoring personalized cancer therapy-an oncologist’s view. Virchows Arch. 2024, 484, 169–179. [Google Scholar] [CrossRef] [PubMed]
  23. Chakravarthi, B.V.; Nepal, S.; Varambally, S. Genomic and Epigenomic Alterations in Cancer. Am. J. Pathol. 2016, 186, 1724–1735. [Google Scholar] [CrossRef] [PubMed]
  24. Chakraborty, S.; Sharma, G.; Karmakar, S.; Banerjee, S. Multi-OMICS approaches in cancer biology: New era in cancer therapy. Biochim. Biophys. Acta Mol. Basis Dis. 2024, 1870, 167120. [Google Scholar] [CrossRef]
  25. Bustin, S.A.; Jellinger, K.A. Advances in Molecular Medicine: Unravelling Disease Complexity and Pioneering Precision Healthcare. Int. J. Mol. Sci. 2023, 24, 14168. [Google Scholar] [CrossRef]
  26. Passaro, A.; Al Bakir, M.; Hamilton, E.G.; Diehn, M.; Andre, F.; Roy-Chowdhuri, S.; Mountzios, G.; Wistuba, I.I.; Swanton, C.; Peters, S. Cancer biomarkers: Emerging trends and clinical implications for personalized treatment. Cell 2024, 187, 1617–1635. [Google Scholar] [CrossRef]
  27. Das, S.; Dey, M.K.; Devireddy, R.; Gartia, M.R. Biomarkers in Cancer Detection, Diagnosis, and Prognosis. Sensors 2023, 24, 37. [Google Scholar] [CrossRef]
  28. Maruvada, P.; Wang, W.; Wagner, P.D.; Srivastava, S. Biomarkers in molecular medicine: Cancer detection and diagnosis. Biotechniques 2005, 38, 9–15. [Google Scholar] [CrossRef]
  29. McNally, C.J.; Ruddock, M.W.; Moore, T.; McKenna, D.J. Biomarkers That Differentiate Benign Prostatic Hyperplasia from Prostate Cancer: A Literature Review. Cancer Manag. Res. 2020, 12, 5225–5241. [Google Scholar] [CrossRef]
  30. Roddam, A.W.; Duffy, M.J.; Hamdy, F.C.; Ward, A.M.; Patnick, J.; Price, C.P.; Rimmer, J.; Sturgeon, C.; White, P.; Allen, N.E. Use of prostate-specific antigen (PSA) isoforms for the detection of prostate cancer in men with a PSA level of 2–10 ng/mL: Systematic review and meta-analysis. Eur. Urol. 2005, 48, 386–399, discussion 398. [Google Scholar] [CrossRef]
  31. Desai, S.; Guddati, A.K. Carcinoembryonic Antigen, Carbohydrate Antigen 19-9, Cancer Antigen 125, Prostate-Specific Antigen and Other Cancer Markers: A Primer on Commonly Used Cancer Markers. World J. Oncol. 2023, 14, 4–14. [Google Scholar] [CrossRef] [PubMed]
  32. Scholler, N.; Urban, N. CA125 in ovarian cancer. Biomark. Med. 2007, 1, 513–523. [Google Scholar] [CrossRef] [PubMed]
  33. Lahoud, R.M.; O’Shea, A.; El-Mouhayyar, C.; Atre, I.D.; Eurboonyanun, K.; Harisinghani, M. Tumour markers and their utility in imaging of abdominal and pelvic malignancies. Clin. Radiol. 2021, 76, 99–107. [Google Scholar] [CrossRef] [PubMed]
  34. Kankanala, V.L.; Mukkamalla, S.K.R. Carcinoembryonic Antigen. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  35. Hanif, H.; Ali, M.J.; Susheela, A.T.; Khan, I.W.; Luna-Cuadros, M.A.; Khan, M.M.; Lau, D.T. Update on the applications and limitations of alpha-fetoprotein for hepatocellular carcinoma. World J. Gastroenterol. 2022, 28, 216–229. [Google Scholar] [CrossRef]
  36. Kawai, M.; Furuhashi, Y.; Kano, T.; Misawa, T.; Nakashima, N.; Hattori, S.; Okamoto, Y.; Kobayashi, I.; Ohta, M.; Arii, Y.; et al. Alpha-fetoprotein in malignant germ cell tumors of the ovary. Gynecol. Oncol. 1990, 39, 160–166. [Google Scholar] [CrossRef]
  37. Turk, A.A.; Wisinski, K.B. PARP inhibitors in breast cancer: Bringing synthetic lethality to the bedside. Cancer 2018, 124, 2498–2506. [Google Scholar] [CrossRef]
  38. Bera, A.; Russ, E.; Karaian, J.; Landa, A.; Radhakrishnan, S.; Subramanian, M.; Hueman, M.; Pollard, H.B.; Hu, H.; Shriver, C.D.; et al. Circulating Cell-free DNA in Serum as a Marker for the Early Detection of Tumor Recurrence in Breast Cancer Patients. Cancer Diagn. Progn. 2022, 2, 285–292. [Google Scholar] [CrossRef]
  39. Riely, G.J.; Politi, K.A.; Miller, V.A.; Pao, W. Update on epidermal growth factor receptor mutations in non-small cell lung cancer. Clin. Cancer Res. 2006, 12, 7232–7241. [Google Scholar] [CrossRef]
  40. Ntzifa, A.; Kotsakis, A.; Georgoulias, V.; Lianidou, E. Detection of EGFR Mutations in Plasma cfDNA and Paired CTCs of NSCLC Patients before and after Osimertinib Therapy Using Crystal Digital PCR. Cancers 2021, 13, 2736. [Google Scholar] [CrossRef]
  41. Shah, R.; Lester, J.F. Tyrosine Kinase Inhibitors for the Treatment of EGFR Mutation-Positive Non-Small-Cell Lung Cancer: A Clash of the Generations. Clin. Lung Cancer 2020, 21, e216–e228. [Google Scholar] [CrossRef]
  42. Dowler Nygaard, A.; Spindler, K.L.; Pallisgaard, N.; Andersen, R.F.; Jakobsen, A. Levels of cell-free DNA and plasma KRAS during treatment of advanced NSCLC. Oncol. Rep. 2014, 31, 969–974. [Google Scholar] [CrossRef] [PubMed]
  43. Takano, S.; Fukasawa, M.; Shindo, H.; Takahashi, E.; Fukasawa, Y.; Kawakami, S.; Hayakawa, H.; Kuratomi, N.; Kadokura, M.; Maekawa, S.; et al. Digital next-generation sequencing of cell-free DNA for pancreatic cancer. JGH Open 2021, 5, 508–516. [Google Scholar] [CrossRef] [PubMed]
  44. Warren, J.D.; Xiong, W.; Bunker, A.M.; Vaughn, C.P.; Furtado, L.V.; Roberts, W.L.; Fang, J.C.; Samowitz, W.S.; Heichman, K.A. Septin 9 methylated DNA is a sensitive and specific blood test for colorectal cancer. BMC Med. 2011, 9, 133. [Google Scholar] [CrossRef]
  45. Lima, A.B.; Dos Reis, M.B.; Matsushita, M.; Dos Reis, M.T.; de Oliveira, M.A.; Reis, R.M.; Guimarães, D.P. Combined SEPT9 and BMP3 methylation in plasma for colorectal cancer early detection and screening in a Brazilian population. Cancer Med. 2023, 12, 15854–15867. [Google Scholar] [CrossRef] [PubMed]
  46. Szylberg, M.; Sokal, P.; Sledzinska, P.; Bebyn, M.; Krajewski, S.; Szylberg, L.; Szylberg, A.; Szylberg, T.; Krystkiewicz, K.; Birski, M.; et al. MGMT Promoter Methylation as a Prognostic Factor in Primary Glioblastoma: A Single-Institution Observational Study. Biomedicines 2022, 10, 2030. [Google Scholar] [CrossRef] [PubMed]
  47. Bagley, S.J.; Till, J.; Abdalla, A.; Sangha, H.K.; Yee, S.S.; Freedman, J.; Black, T.A.; Hussain, J.; Binder, Z.A.; Brem, S.; et al. Association of plasma cell-free DNA with survival in patients with IDH wild-type glioblastoma. Neurooncol Adv. 2021, 3, vdab011. [Google Scholar] [CrossRef]
  48. Fang, X.; Yu, S.; Bast, R.C.; Liu, S.; Xu, H.J.; Hu, S.X.; LaPushin, R.; Claret, F.X.; Aggarwal, B.B.; Lu, Y.; et al. Mechanisms for lysophosphatidic acid-induced cytokine production in ovarian cancer cells. J. Biol. Chem. 2004, 279, 9653–9661. [Google Scholar] [CrossRef]
  49. Balijepalli, P.; Sitton, C.C.; Meier, K.E. Lysophosphatidic Acid Signaling in Cancer Cells: What Makes LPA So Special? Cells 2021, 10, 2059. [Google Scholar] [CrossRef]
  50. Xu, Y.; Shen, Z.; Wiper, D.W.; Wu, M.; Morton, R.E.; Elson, P.; Kennedy, A.W.; Belinson, J.; Markman, M.; Casey, G. Lysophosphatidic acid as a potential biomarker for ovarian and other gynecologic cancers. JAMA 1998, 280, 719–723. [Google Scholar] [CrossRef]
  51. Harewood, R.; Rothwell, J.A.; Besevic, J.; Viallon, V.; Achaintre, D.; Gicquiau, A.; Rinaldi, S.; Wedekind, R.; Prehn, C.; Adamski, J.; et al. Association between pre-diagnostic circulating lipid metabolites and colorectal cancer risk: A nested case-control study in the European Prospective Investigation into Cancer and Nutrition (EPIC). EBioMedicine 2024, 101, 105024. [Google Scholar] [CrossRef]
  52. Ide, Y.; Waki, M.; Hayasaka, T.; Nishio, T.; Morita, Y.; Tanaka, H.; Sasaki, T.; Koizumi, K.; Matsunuma, R.; Hosokawa, Y.; et al. Human breast cancer tissues contain abundant phosphatidylcholine(36ratio1) with high stearoyl-CoA desaturase-1 expression. PLoS ONE 2013, 8, e61204. [Google Scholar] [CrossRef]
  53. Sakakima, Y.; Hayakawa, A.; Nakao, A. Phosphatidylcholine induces growth inhibition of hepatic cancer by apoptosis via death ligands. Hepatogastroenterology 2009, 56, 481–484. [Google Scholar] [PubMed]
  54. Kühn, T.; Floegel, A.; Sookthai, D.; Johnson, T.; Rolle-Kampczyk, U.; Otto, W.; von Bergen, M.; Boeing, H.; Kaaks, R. Higher plasma levels of lysophosphatidylcholine 18:0 are related to a lower risk of common cancers in a prospective metabolomics study. BMC Med. 2016, 14, 13. [Google Scholar] [CrossRef]
  55. Nagahashi, M.; Abe, M.; Sakimura, K.; Takabe, K.; Wakai, T. The role of sphingosine-1-phosphate in inflammation and cancer progression. Cancer Sci. 2018, 109, 3671–3678. [Google Scholar] [CrossRef] [PubMed]
  56. Fan, Q.; Cheng, Y.; Chang, H.M.; Deguchi, M.; Hsueh, A.J.; Leung, P.C.K. Sphingosine-1-phosphate promotes ovarian cancer cell proliferation by disrupting Hippo signaling. Oncotarget 2017, 8, 27166–27176. [Google Scholar] [CrossRef]
  57. Wang, D.; Zhao, Z.; Caperell-Grant, A.; Yang, G.; Mok, S.C.; Liu, J.; Bigsby, R.M.; Xu, Y. S1P differentially regulates migration of human ovarian cancer and human ovarian surface epithelial cells. Mol. Cancer Ther. 2008, 7, 1993–2002. [Google Scholar] [CrossRef]
  58. Wajapeyee, N.; Beamon, T.C.; Gupta, R. Roles and therapeutic targeting of ceramide metabolism in cancer. Mol. Metab. 2024, 83, 101936. [Google Scholar] [CrossRef]
  59. Moro, K.; Kawaguchi, T.; Tsuchida, J.; Gabriel, E.; Qi, Q.; Yan, L.; Wakai, T.; Takabe, K.; Nagahashi, M. Ceramide species are elevated in human breast cancer and are associated with less aggressiveness. Oncotarget 2018, 9, 19874–19890. [Google Scholar] [CrossRef]
  60. Nelson, E.R.; Wardell, S.E.; Jasper, J.S.; Park, S.; Suchindran, S.; Howe, M.K.; Carver, N.J.; Pillai, R.V.; Sullivan, P.M.; Sondhi, V.; et al. 27-Hydroxycholesterol links hypercholesterolemia and breast cancer pathophysiology. Science 2013, 342, 1094–1098. [Google Scholar] [CrossRef]
  61. Kimbung, S.; Chang, C.Y.; Bendahl, P.O.; Dubois, L.; Thompson, J.W.; McDonnell, D.P.; Borgquist, S. Impact of 27-hydroxylase (CYP27A1) and 27-hydroxycholesterol in breast cancer. Endocr. Relat. Cancer 2017, 24, 339–349. [Google Scholar] [CrossRef]
  62. Yue, S.; Li, J.; Lee, S.Y.; Lee, H.J.; Shao, T.; Song, B.; Cheng, L.; Masterson, T.A.; Liu, X.; Ratliff, T.L.; et al. Cholesteryl ester accumulation induced by PTEN loss and PI3K/AKT activation underlies human prostate cancer aggressiveness. Cell Metab. 2014, 19, 393–406. [Google Scholar] [CrossRef] [PubMed]
  63. Guo, X.; Zhou, S.; Yang, Z.; Li, Z.A.; Hu, W.; Dai, L.; Liang, W.; Wang, X. Cholesterol metabolism and its implication in glioblastoma therapy. J. Cancer 2022, 13, 1745–1757. [Google Scholar] [CrossRef] [PubMed]
  64. Dahm, C.C.; Gorst-Rasmussen, A.; Crowe, F.L.; Roswall, N.; Tjonneland, A.; Drogan, D.; Boeing, H.; Teucher, B.; Kaaks, R.; Adarakis, G.; et al. Fatty acid patterns and risk of prostate cancer in a case-control study nested within the European Prospective Investigation into Cancer and Nutrition. Am. J. Clin. Nutr. 2012, 96, 1354–1361. [Google Scholar] [CrossRef]
  65. Marcial-Medina, C.; Ordonez-Moreno, A.; Gonzalez-Reyes, C.; Cortes-Reynosa, P.; Perez Salazar, E. Oleic acid induces migration through a FFAR1/4, EGFR and AKT-dependent pathway in breast cancer cells. Endocr. Connect. 2019, 8, 252–265. [Google Scholar] [CrossRef]
  66. Staehr, P.; Hother-Nielsen, O.; Landau, B.R.; Chandramouli, V.; Holst, J.J.; Beck-Nielsen, H. Effects of free fatty acids per se on glucose production, gluconeogenesis, and glycogenolysis. Diabetes 2003, 52, 260–267. [Google Scholar] [CrossRef]
  67. Landegren, U.; Hammond, M. Cancer diagnostics based on plasma protein biomarkers: Hard times but great expectations. Mol. Oncol. 2021, 15, 1715–1726. [Google Scholar] [CrossRef]
  68. Quraish, R.U.; Hirahata, T.; Quraish, A.U.; Ul Quraish, S. An Overview: Genetic Tumor Markers for Early Detection and Current Gene Therapy Strategies. Cancer Inform. 2023, 22, 11769351221150772. [Google Scholar] [CrossRef]
  69. Barili, V.; Ambrosini, E.; Bortesi, B.; Minari, R.; De Sensi, E.; Cannizzaro, I.R.; Taiani, A.; Michiara, M.; Sikokis, A.; Boggiani, D.; et al. Genetic Basis of Breast and Ovarian Cancer: Approaches and Lessons Learnt from Three Decades of Inherited Predisposition Testing. Genes 2024, 15, 219. [Google Scholar] [CrossRef]
  70. Misale, S.; Di Nicolantonio, F.; Sartore-Bianchi, A.; Siena, S.; Bardelli, A. Resistance to anti-EGFR therapy in colorectal cancer: From heterogeneity to convergent evolution. Cancer Discov. 2014, 4, 1269–1280. [Google Scholar] [CrossRef]
  71. Zhou, Y.; Tao, L.; Qiu, J.; Xu, J.; Yang, X.; Zhang, Y.; Tian, X.; Guan, X.; Cen, X.; Zhao, Y. Tumor biomarkers for diagnosis, prognosis and targeted therapy. Signal Transduct. Target. Ther. 2024, 9, 132. [Google Scholar] [CrossRef]
  72. Kaminska, K.; Nalejska, E.; Kubiak, M.; Wojtysiak, J.; Zolna, L.; Kowalewski, J.; Lewandowska, M.A. Prognostic and Predictive Epigenetic Biomarkers in Oncology. Mol. Diagn. Ther. 2019, 23, 83–95. [Google Scholar] [CrossRef] [PubMed]
  73. Gao, J.; Shi, W.; Wang, J.; Guan, C.; Dong, Q.; Sheng, J.; Zou, X.; Xu, Z.; Ge, Y.; Yang, C.; et al. Research progress and applications of epigenetic biomarkers in cancer. Front. Pharmacol. 2024, 15, 1308309. [Google Scholar] [CrossRef] [PubMed]
  74. Hanna, M.; Dey, N.; Grady, W.M. Emerging Tests for Noninvasive Colorectal Cancer Screening. Clin. Gastroenterol. Hepatol. 2023, 21, 604–616. [Google Scholar] [CrossRef] [PubMed]
  75. Lahtz, C.; Pfeifer, G.P. Epigenetic changes of DNA repair genes in cancer. J. Mol. Cell Biol. 2011, 3, 51–58. [Google Scholar] [CrossRef]
  76. Butler, L.M.; Perone, Y.; Dehairs, J.; Lupien, L.E.; de Laat, V.; Talebi, A.; Loda, M.; Kinlaw, W.B.; Swinnen, J.V. Lipids and cancer: Emerging roles in pathogenesis, diagnosis and therapeutic intervention. Adv. Drug Deliv. Rev. 2020, 159, 245–293. [Google Scholar] [CrossRef]
  77. Martin-Perez, M.; Urdiroz-Urricelqui, U.; Bigas, C.; Benitah, S.A. The role of lipids in cancer progression and metastasis. Cell Metab. 2022, 34, 1675–1699. [Google Scholar] [CrossRef]
  78. Fu, Y.; Zou, T.; Shen, X.; Nelson, P.J.; Li, J.; Wu, C.; Yang, J.; Zheng, Y.; Bruns, C.; Zhao, Y.; et al. Lipid metabolism in cancer progression and therapeutic strategies. MedComm 2021, 2, 27–59. [Google Scholar] [CrossRef]
  79. Szlasa, W.; Zendran, I.; Zalesinska, A.; Tarek, M.; Kulbacka, J. Lipid composition of the cancer cell membrane. J. Bioenerg. Biomembr. 2020, 52, 321–342. [Google Scholar] [CrossRef]
  80. Geraldo, L.H.M.; Spohr, T.; Amaral, R.F.D.; Fonseca, A.; Garcia, C.; Mendes, F.A.; Freitas, C.; dosSantos, M.F.; Lima, F.R.S. Role of lysophosphatidic acid and its receptors in health and disease: Novel therapeutic strategies. Signal Transduct. Target. Ther. 2021, 6, 45. [Google Scholar] [CrossRef]
  81. van der Veen, J.N.; Kennelly, J.P.; Wan, S.; Vance, J.E.; Vance, D.E.; Jacobs, R.L. The critical role of phosphatidylcholine and phosphatidylethanolamine metabolism in health and disease. Biochim. Biophys. Acta Biomembr. 2017, 1859, 1558–1572. [Google Scholar] [CrossRef]
  82. Li, R.Z.; Wang, X.R.; Wang, J.; Xie, C.; Wang, X.X.; Pan, H.D.; Meng, W.Y.; Liang, T.L.; Li, J.X.; Yan, P.Y.; et al. The key role of sphingolipid metabolism in cancer: New therapeutic targets, diagnostic and prognostic values, and anti-tumor immunotherapy resistance. Front. Oncol. 2022, 12, 941643. [Google Scholar] [CrossRef] [PubMed]
  83. Segui, B.; Andrieu-Abadie, N.; Jaffrezou, J.P.; Benoist, H.; Levade, T. Sphingolipids as modulators of cancer cell death: Potential therapeutic targets. Biochim. Biophys. Acta 2006, 1758, 2104–2120. [Google Scholar] [CrossRef] [PubMed]
  84. Hait, N.C.; Maiti, A. The Role of Sphingosine-1-Phosphate and Ceramide-1-Phosphate in Inflammation and Cancer. Mediators Inflamm. 2017, 2017, 4806541. [Google Scholar] [CrossRef] [PubMed]
  85. Pilatova, M.B.; Solarova, Z.; Mezencev, R.; Solar, P. Ceramides and their roles in programmed cell death. Adv. Med. Sci. 2023, 68, 417–425. [Google Scholar] [CrossRef]
  86. Vona, R.; Iessi, E.; Matarrese, P. Role of Cholesterol and Lipid Rafts in Cancer Signaling: A Promising Therapeutic Opportunity? Front. Cell Dev. Biol. 2021, 9, 622908. [Google Scholar] [CrossRef]
  87. Xiao, M.; Xu, J.; Wang, W.; Zhang, B.; Liu, J.; Li, J.; Xu, H.; Zhao, Y.; Yu, X.; Shi, S. Functional significance of cholesterol metabolism in cancer: From threat to treatment. Exp. Mol. Med. 2023, 55, 1982–1995. [Google Scholar] [CrossRef]
  88. Liu, Z.; Gomez, C.R.; Espinoza, I.; Le, T.P.T.; Shenoy, V.; Zhou, X. Correlation of cholesteryl ester metabolism to pathogenesis, progression and disparities in colorectal Cancer. Lipids Health Dis. 2022, 21, 22. [Google Scholar] [CrossRef]
  89. Mallick, R.; Bhowmik, P.; Duttaroy, A.K. Targeting fatty acid uptake and metabolism in cancer cells: A promising strategy for cancer treatment. Biomed. Pharmacother. 2023, 167, 115591. [Google Scholar] [CrossRef]
  90. Corn, K.C.; Windham, M.A.; Rafat, M. Lipids in the tumor microenvironment: From cancer progression to treatment. Prog. Lipid Res. 2020, 80, 101055. [Google Scholar] [CrossRef]
  91. Zhang, Y.; He, C.; Qiu, L.; Wang, Y.; Zhang, L.; Qin, X.; Liu, Y.; Zhang, D.; Li, Z. Serum unsaturated free Fatty acids: Potential biomarkers for early detection and disease progression monitoring of non-small cell lung cancer. J. Cancer 2014, 5, 706–714. [Google Scholar] [CrossRef]
  92. Loke, S.Y.; Lee, A.S.G. The future of blood-based biomarkers for the early detection of breast cancer. Eur. J. Cancer 2018, 92, 54–68. [Google Scholar] [CrossRef] [PubMed]
  93. Zanoaga, O.; Jurj, A.; Raduly, L.; Cojocneanu-Petric, R.; Fuentes-Mattei, E.; Wu, O.; Braicu, C.; Gherman, C.D.; Berindan-Neagoe, I. Implications of dietary omega-3 and omega-6 polyunsaturated fatty acids in breast cancer. Exp. Ther. Med. 2018, 15, 1167–1176. [Google Scholar] [CrossRef] [PubMed]
  94. Huerta-Yepez, S.; Tirado-Rodriguez, A.B.; Hankinson, O. Role of diets rich in omega-3 and omega-6 in the development of cancer. Bol. Med. Hosp. Infant. Mex. 2016, 73, 446–456. [Google Scholar] [CrossRef] [PubMed]
  95. Moloudizargari, M.; Mortaz, E.; Asghari, M.H.; Adcock, I.M.; Redegeld, F.A.; Garssen, J. Effects of the polyunsaturated fatty acids, EPA and DHA, on hematological malignancies: A systematic review. Oncotarget 2018, 9, 11858–11875. [Google Scholar] [CrossRef]
  96. Borin, T.F.; Angara, K.; Rashid, M.H.; Achyut, B.R.; Arbab, A.S. Arachidonic Acid Metabolite as a Novel Therapeutic Target in Breast Cancer Metastasis. Int. J. Mol. Sci. 2017, 18, 2661. [Google Scholar] [CrossRef]
  97. Pavlova, N.N.; Thompson, C.B. The Emerging Hallmarks of Cancer Metabolism. Cell Metab. 2016, 23, 27–47. [Google Scholar] [CrossRef]
  98. Pavlova, N.N.; Zhu, J.; Thompson, C.B. The hallmarks of cancer metabolism: Still emerging. Cell Metab. 2022, 34, 355–377. [Google Scholar] [CrossRef]
  99. Park, J.; Shin, Y.; Kim, T.H.; Kim, D.H.; Lee, A. Plasma metabolites as possible biomarkers for diagnosis of breast cancer. PLoS ONE 2019, 14, e0225129. [Google Scholar] [CrossRef]
  100. Saorin, A.; Di Gregorio, E.; Miolo, G.; Steffan, A.; Corona, G. Emerging Role of Metabolomics in Ovarian Cancer Diagnosis. Metabolites 2020, 10, 419. [Google Scholar] [CrossRef]
  101. Al-Daffaie, F.M.; Al-Mudhafar, S.F.; Alhomsi, A.; Tarazi, H.; Almehdi, A.M.; El-Huneidi, W.; Abu-Gharbieh, E.; Bustanji, Y.; Alqudah, M.A.Y.; Abuhelwa, A.Y.; et al. Metabolomics and Proteomics in Prostate Cancer Research: Overview, Analytical Techniques, Data Analysis, and Recent Clinical Applications. Int. J. Mol. Sci. 2024, 25, 5071. [Google Scholar] [CrossRef]
  102. Kdadra, M.; Höckner, S.; Leung, H.; Kremer, W.; Schiffer, E. Metabolomics Biomarkers of Prostate Cancer: A Systematic Review. Diagnostics 2019, 9, 21. [Google Scholar] [CrossRef] [PubMed]
  103. Luo, X.; Liu, J.; Wang, H.; Lu, H. Metabolomics identified new biomarkers for the precise diagnosis of pancreatic cancer and associated tissue metastasis. Pharmacol. Res. 2020, 156, 104805. [Google Scholar] [CrossRef] [PubMed]
  104. Skubisz, K.; Dąbkowski, K.; Samborowska, E.; Starzyńska, T.; Deskur, A.; Ambrozkiewicz, F.; Karczmarski, J.; Radkiewicz, M.; Kusnierz, K.; Kos-Kudła, B.; et al. Serum Metabolite Biomarkers for Pancreatic Tumors: Neuroendocrine and Pancreatic Ductal Adenocarcinomas-A Preliminary Study. Cancers 2023, 15, 3242. [Google Scholar] [CrossRef]
  105. Huang, S.; Guo, Y.; Li, Z.; Zhang, Y.; Zhou, T.; You, W.; Pan, K.; Li, W. A systematic review of metabolomic profiling of gastric cancer and esophageal cancer. Cancer Biol. Med. 2020, 17, 181–198. [Google Scholar] [CrossRef] [PubMed]
  106. Tan, B.; Qiu, Y.; Zou, X.; Chen, T.; Xie, G.; Cheng, Y.; Dong, T.; Zhao, L.; Feng, B.; Hu, X.; et al. Metabonomics identifies serum metabolite markers of colorectal cancer. J. Proteome Res. 2013, 12, 3000–3009. [Google Scholar] [CrossRef] [PubMed]
  107. Ossoliński, K.; Ruman, T.; Copié, V.; Tripet, B.P.; Nogueira, L.B.; Nogueira, K.; Kołodziej, A.; Płaza-Altamer, A.; Ossolińska, A.; Ossoliński, T.; et al. Metabolomic and elemental profiling of blood serum in bladder cancer. J. Pharm. Anal. 2022, 12, 889–900. [Google Scholar] [CrossRef]
  108. Wang, J.H.; Chen, W.L.; Li, J.M.; Wu, S.F.; Chen, T.L.; Zhu, Y.M.; Zhang, W.N.; Li, Y.; Qiu, Y.P.; Zhao, A.H.; et al. Prognostic significance of 2-hydroxyglutarate levels in acute myeloid leukemia in China. Proc. Natl. Acad. Sci. USA 2013, 110, 17017–17022. [Google Scholar] [CrossRef]
  109. Coelho, M.; Raposo, L.; Goodfellow, B.J.; Atzori, L.; Jones, J.; Manadas, B. The Potential of Metabolomics in the Diagnosis of Thyroid Cancer. Int. J. Mol. Sci. 2020, 21, 5272. [Google Scholar] [CrossRef]
  110. Xu, H.; Liu, R.; He, B.; Bi, C.W.; Bi, K.; Li, Q. Polyamine Metabolites Profiling for Characterization of Lung and Liver Cancer Using an LC-Tandem MS Method with Multiple Statistical Data Mining Strategies: Discovering Potential Cancer Biomarkers in Human Plasma and Urine. Molecules 2016, 21, 1040. [Google Scholar] [CrossRef]
  111. Najafi, S.; Khatami, S.H.; Khorsand, M.; Jamali, Z.; Shabaninejad, Z.; Moazamfard, M.; Majidpoor, J.; Aghaei Zarch, S.M.; Movahedpour, A. Long non-coding RNAs (lncRNAs); roles in tumorigenesis and potentials as biomarkers in cancer diagnosis. Exp. Cell Res. 2022, 418, 113294. [Google Scholar] [CrossRef]
  112. Gupta, R.A.; Shah, N.; Wang, K.C.; Kim, J.; Horlings, H.M.; Wong, D.J.; Tsai, M.C.; Hung, T.; Argani, P.; Rinn, J.L.; et al. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 2010, 464, 1071–1076. [Google Scholar] [CrossRef] [PubMed]
  113. Lu, K.H.; Li, W.; Liu, X.H.; Sun, M.; Zhang, M.L.; Wu, W.Q.; Xie, W.P.; Hou, Y.Y. Long non-coding RNA MEG3 inhibits NSCLC cells proliferation and induces apoptosis by affecting p53 expression. BMC Cancer 2013, 13, 461. [Google Scholar] [CrossRef] [PubMed]
  114. Peng, Y.; Croce, C.M. The role of MicroRNAs in human cancer. Signal Transduct. Target. Ther. 2016, 1, 15004. [Google Scholar] [CrossRef] [PubMed]
  115. Silva, J.M.; Boczek, N.J.; Berres, M.W.; Ma, X.; Smith, D.I. LSINCT5 is over expressed in breast and ovarian cancer and affects cellular proliferation. RNA Biol. 2011, 8, 496–505. [Google Scholar] [CrossRef]
  116. Li, H.; Zhu, L.; Xu, L.; Qin, K.; Liu, C.; Yu, Y.; Su, D.; Wu, K.; Sheng, Y. Long noncoding RNA linc00617 exhibits oncogenic activity in breast cancer. Mol. Carcinog. 2017, 56, 3–17. [Google Scholar] [CrossRef] [PubMed]
  117. Xu, N.; Chen, F.; Wang, F.; Lu, X.; Wang, X.; Lv, M.; Lu, C. Clinical significance of high expression of circulating serum lncRNA RP11-445H22.4 in breast cancer patients: A Chinese population-based study. Tumour Biol. 2015, 36, 7659–7665. [Google Scholar] [CrossRef]
  118. Iacoangeli, A.; Lin, Y.; Morley, E.J.; Muslimov, I.A.; Bianchi, R.; Reilly, J.; Weedon, J.; Diallo, R.; Böcker, W.; Tiedge, H. BC200 RNA in invasive and preinvasive breast cancer. Carcinogenesis 2004, 25, 2125–2133. [Google Scholar] [CrossRef]
  119. Nie, W.; Ge, H.J.; Yang, X.Q.; Sun, X.; Huang, H.; Tao, X.; Chen, W.S.; Li, B. LncRNA-UCA1 exerts oncogenic functions in non-small cell lung cancer by targeting miR-193a-3p. Cancer Lett. 2016, 371, 99–106. [Google Scholar] [CrossRef]
  120. Yan, R.; Wang, K.; Peng, R.; Wang, S.; Cao, J.; Wang, P.; Song, C. Genetic variants in lncRNA SRA and risk of breast cancer. Oncotarget 2016, 7, 22486–22496. [Google Scholar] [CrossRef]
  121. Zhao, H.; Shen, J.; Medico, L.; Wang, D.; Ambrosone, C.B.; Liu, S. A pilot study of circulating miRNAs as potential biomarkers of early stage breast cancer. PLoS ONE 2010, 5, e13735. [Google Scholar] [CrossRef]
  122. Wang, F.; Zheng, Z.; Guo, J.; Ding, X. Correlation and quantitation of microRNA aberrant expression in tissues and sera from patients with breast tumor. Gynecol. Oncol. 2010, 119, 586–593. [Google Scholar] [CrossRef] [PubMed]
  123. Roth, C.; Rack, B.; Müller, V.; Janni, W.; Pantel, K.; Schwarzenbach, H. Circulating microRNAs as blood-based markers for patients with primary and metastatic breast cancer. Breast Cancer Res. 2010, 12, R90. [Google Scholar] [CrossRef] [PubMed]
  124. Heneghan, H.M.; Miller, N.; Kelly, R.; Newell, J.; Kerin, M.J. Systemic miRNA-195 differentiates breast cancer from other malignancies and is a potential biomarker for detecting noninvasive and early stage disease. Oncologist 2010, 15, 673–682. [Google Scholar] [CrossRef] [PubMed]
  125. Heneghan, H.M.; Miller, N.; Lowery, A.J.; Sweeney, K.J.; Newell, J.; Kerin, M.J. Circulating microRNAs as novel minimally invasive biomarkers for breast cancer. Ann. Surg. 2010, 251, 499–505. [Google Scholar] [CrossRef]
  126. Wang, P.; Ren, Z.; Sun, P. Overexpression of the long non-coding RNA MEG3 impairs in vitro glioma cell proliferation. J. Cell Biochem. 2012, 113, 1868–1874. [Google Scholar] [CrossRef]
  127. Zhao, X.; Wang, P.; Liu, J.; Zheng, J.; Liu, Y.; Chen, J.; Xue, Y. Gas5 Exerts Tumor-suppressive Functions in Human Glioma Cells by Targeting miR-222. Mol. Ther. 2015, 23, 1899–1911. [Google Scholar] [CrossRef]
  128. Qin, X.; Yao, J.; Geng, P.; Fu, X.; Xue, J.; Zhang, Z. LncRNA TSLC1-AS1 is a novel tumor suppressor in glioma. Int. J. Clin. Exp. Pathol. 2014, 7, 3065–3072. [Google Scholar]
  129. Yao, J.; Zhou, B.; Zhang, J.; Geng, P.; Liu, K.; Zhu, Y.; Zhu, W. A new tumor suppressor LncRNA ADAMTS9-AS2 is regulated by DNMT1 and inhibits migration of glioma cells. Tumour Biol. 2014, 35, 7935–7944. [Google Scholar] [CrossRef]
  130. Yue, H.; Zhu, J.; Xie, S.; Li, F.; Xu, Q. MDC1-AS, an antisense long noncoding RNA, regulates cell proliferation of glioma. Biomed. Pharmacother. 2016, 81, 203–209. [Google Scholar] [CrossRef]
  131. Li, J.; Zhang, M.; An, G.; Ma, Q. LncRNA TUG1 acts as a tumor suppressor in human glioma by promoting cell apoptosis. Exp Biol. Med. 2016, 241, 644–649. [Google Scholar] [CrossRef]
  132. Feng, S.; Yao, J.; Chen, Y.; Geng, P.; Zhang, H.; Ma, X.; Zhao, J.; Yu, X. Expression and Functional Role of Reprogramming-Related Long Noncoding RNA (lincRNA-ROR) in Glioma. J. Mol. Neurosci. 2015, 56, 623–630. [Google Scholar] [CrossRef] [PubMed]
  133. Wang, P.; Liu, Y.H.; Yao, Y.L.; Li, Z.; Li, Z.Q.; Ma, J.; Xue, Y.X. Long non-coding RNA CASC2 suppresses malignancy in human gliomas by miR-21. Cell Signal 2015, 27, 275–282. [Google Scholar] [CrossRef] [PubMed]
  134. Yao, Y.; Ma, J.; Xue, Y.; Wang, P.; Li, Z.; Liu, J.; Chen, L.; Xi, Z.; Teng, H.; Wang, Z.; et al. Knockdown of long non-coding RNA XIST exerts tumor-suppressive functions in human glioblastoma stem cells by up-regulating miR-152. Cancer Lett. 2015, 359, 75–86. [Google Scholar] [CrossRef]
  135. Wang, Y.; Wang, Y.; Li, J.; Zhang, Y.; Yin, H.; Han, B. CRNDE, a long-noncoding RNA, promotes glioma cell growth and invasion through mTOR signaling. Cancer Lett. 2015, 367, 122–128. [Google Scholar] [CrossRef]
  136. Ma, J.; Wang, P.; Yao, Y.; Liu, Y.; Li, Z.; Liu, X.; Li, Z.; Zhao, X.; Xi, Z.; Teng, H.; et al. Knockdown of long non-coding RNA MALAT1 increases the blood-tumor barrier permeability by up-regulating miR-140. Biochim. Biophys. Acta 2016, 1859, 324–338. [Google Scholar] [CrossRef]
  137. Zhang, J.X.; Han, L.; Bao, Z.S.; Wang, Y.Y.; Chen, L.Y.; Yan, W.; Yu, S.Z.; Pu, P.Y.; Liu, N.; You, Y.P.; et al. HOTAIR, a cell cycle-associated long noncoding RNA and a strong predictor of survival, is preferentially expressed in classical and mesenchymal glioma. Neuro Oncol. 2013, 15, 1595–1603. [Google Scholar] [CrossRef]
  138. Ke, J.; Yao, Y.L.; Zheng, J.; Wang, P.; Liu, Y.H.; Ma, J.; Li, Z.; Liu, X.B.; Li, Z.Q.; Wang, Z.H.; et al. Knockdown of long non-coding RNA HOTAIR inhibits malignant biological behaviors of human glioma cells via modulation of miR-326. Oncotarget 2015, 6, 21934–21949. [Google Scholar] [CrossRef]
  139. Wang, Q.; Zhang, J.; Liu, Y.; Zhang, W.; Zhou, J.; Duan, R.; Pu, P.; Kang, C.; Han, L. A novel cell cycle-associated lncRNA, HOXA11-AS, is transcribed from the 5-prime end of the HOXA transcript and is a biomarker of progression in glioma. Cancer Lett. 2016, 373, 251–259. [Google Scholar] [CrossRef]
  140. Guo, H.; Wu, L.; Yang, Q.; Ye, M.; Zhu, X. Functional linc-POU3F3 is overexpressed and contributes to tumorigenesis in glioma. Gene 2015, 554, 114–119. [Google Scholar] [CrossRef]
  141. Ma, C.C.; Xiong, Z.; Zhu, G.N.; Wang, C.; Zong, G.; Wang, H.L.; Bian, E.B.; Zhao, B. Long non-coding RNA ATB promotes glioma malignancy by negatively regulating miR-200a. J. Exp. Clin. Cancer Res. 2016, 35, 90. [Google Scholar] [CrossRef]
  142. Hu, L.; Lv, Q.L.; Chen, S.H.; Sun, B.; Qu, Q.; Cheng, L.; Guo, Y.; Zhou, H.H.; Fan, L. Up-Regulation of Long Non-Coding RNA AB073614 Predicts a Poor Prognosis in Patients with Glioma. Int. J. Environ. Res. Public Health 2016, 13, 433. [Google Scholar] [CrossRef] [PubMed]
  143. Liu, H.; Lv, Z.; Guo, E. Knockdown of long noncoding RNA SPRY4-IT1 suppresses glioma cell proliferation, metastasis and epithelial-mesenchymal transition. Int. J. Clin. Exp. Pathol. 2015, 8, 9140–9146. [Google Scholar] [PubMed]
  144. Shi, Y.; Wang, Y.; Luan, W.; Wang, P.; Tao, T.; Zhang, J.; Qian, J.; Liu, N.; You, Y. Long non-coding RNA H19 promotes glioma cell invasion by deriving miR-675. PLoS ONE 2014, 9, e86295. [Google Scholar] [CrossRef] [PubMed]
  145. Zhou, Q.; Liu, J.; Quan, J.; Liu, W.; Tan, H.; Li, W. MicroRNAs as potential biomarkers for the diagnosis of glioma: A systematic review and meta-analysis. Cancer Sci. 2018, 109, 2651–2659. [Google Scholar] [CrossRef] [PubMed]
  146. Takahashi, Y.; Sawada, G.; Kurashige, J.; Uchi, R.; Matsumura, T.; Ueo, H.; Takano, Y.; Eguchi, H.; Sudo, T.; Sugimachi, K.; et al. Amplification of PVT-1 is involved in poor prognosis via apoptosis inhibition in colorectal cancers. Br. J. Cancer 2014, 110, 164–171. [Google Scholar] [CrossRef]
  147. Ding, J.; Lu, B.; Wang, J.; Wang, J.; Shi, Y.; Lian, Y.; Zhu, Y.; Wang, J.; Fan, Y.; Wang, Z.; et al. Long non-coding RNA Loc554202 induces apoptosis in colorectal cancer cells via the caspase cleavage cascades. J. Exp. Clin. Cancer Res. 2015, 34, 100. [Google Scholar] [CrossRef]
  148. Yang, W.; Ning, N.; Jin, X. The lncRNA H19 Promotes Cell Proliferation by Competitively Binding to miR-200a and Derepressing β-Catenin Expression in Colorectal Cancer. Biomed. Res. Int. 2017, 2017, 2767484. [Google Scholar] [CrossRef]
  149. Han, X.; Wang, L.; Ning, Y.; Li, S.; Wang, Z. Long non-coding RNA AFAP1-AS1 facilitates tumor growth and promotes metastasis in colorectal cancer. Biol. Res. 2016, 49, 36. [Google Scholar] [CrossRef]
  150. Yang, M.H.; Hu, Z.Y.; Xu, C.; Xie, L.Y.; Wang, X.Y.; Chen, S.Y.; Li, Z.G. MALAT1 promotes colorectal cancer cell proliferation/migration/invasion via PRKA kinase anchor protein 9. Biochim. Biophys. Acta 2015, 1852, 166–174. [Google Scholar] [CrossRef]
  151. Xiang, J.F.; Yin, Q.F.; Chen, T.; Zhang, Y.; Zhang, X.O.; Wu, Z.; Zhang, S.; Wang, H.B.; Ge, J.; Lu, X.; et al. Human colorectal cancer-specific CCAT1-L lncRNA regulates long-range chromatin interactions at the MYC locus. Cell Res. 2014, 24, 513–531. [Google Scholar] [CrossRef]
  152. Ge, X.; Chen, Y.; Liao, X.; Liu, D.; Li, F.; Ruan, H.; Jia, W. Overexpression of long noncoding RNA PCAT-1 is a novel biomarker of poor prognosis in patients with colorectal cancer. Med. Oncol. 2013, 30, 588. [Google Scholar] [CrossRef] [PubMed]
  153. Huang, Z.; Huang, D.; Ni, S.; Peng, Z.; Sheng, W.; Du, X. Plasma microRNAs are promising novel biomarkers for early detection of colorectal cancer. Int. J. Cancer 2010, 127, 118–126. [Google Scholar] [CrossRef] [PubMed]
  154. Cheng, H.; Zhang, L.; Cogdell, D.E.; Zheng, H.; Schetter, A.J.; Nykter, M.; Harris, C.C.; Chen, K.; Hamilton, S.R.; Zhang, W. Circulating plasma MiR-141 is a novel biomarker for metastatic colon cancer and predicts poor prognosis. PLoS ONE 2011, 6, e17745. [Google Scholar] [CrossRef] [PubMed]
  155. Pu, X.X.; Huang, G.L.; Guo, H.Q.; Guo, C.C.; Li, H.; Ye, S.; Ling, S.; Jiang, L.; Tian, Y.; Lin, T.Y. Circulating miR-221 directly amplified from plasma is a potential diagnostic and prognostic marker of colorectal cancer and is correlated with p53 expression. J. Gastroenterol. Hepatol. 2010, 25, 1674–1680. [Google Scholar] [CrossRef]
  156. Poliseno, L.; Salmena, L.; Zhang, J.; Carver, B.; Haveman, W.J.; Pandolfi, P.P. A coding-independent function of gene and pseudogene mRNAs regulates tumour biology. Nature 2010, 465, 1033–1038. [Google Scholar] [CrossRef]
  157. Merola, R.; Tomao, L.; Antenucci, A.; Sperduti, I.; Sentinelli, S.; Masi, S.; Mandoj, C.; Orlandi, G.; Papalia, R.; Guaglianone, S.; et al. PCA3 in prostate cancer and tumor aggressiveness detection on 407 high-risk patients: A National Cancer Institute experience. J. Exp. Clin. Cancer Res. 2015, 34, 15. [Google Scholar] [CrossRef]
  158. Ylipää, A.; Kivinummi, K.; Kohvakka, A.; Annala, M.; Latonen, L.; Scaravilli, M.; Kartasalo, K.; Leppänen, S.P.; Karakurt, S.; Seppälä, J.; et al. Transcriptome Sequencing Reveals PCAT5 as a Novel ERG-Regulated Long Noncoding RNA in Prostate Cancer. Cancer Res. 2015, 75, 4026–4031. [Google Scholar] [CrossRef]
  159. Crea, F.; Watahiki, A.; Quagliata, L.; Xue, H.; Pikor, L.; Parolia, A.; Wang, Y.; Lin, D.; Lam, W.L.; Farrar, W.L.; et al. Identification of a long non-coding RNA as a novel biomarker and potential therapeutic target for metastatic prostate cancer. Oncotarget 2014, 5, 764–774. [Google Scholar] [CrossRef]
  160. Chung, S.; Nakagawa, H.; Uemura, M.; Piao, L.; Ashikawa, K.; Hosono, N.; Takata, R.; Akamatsu, S.; Kawaguchi, T.; Morizono, T.; et al. Association of a novel long non-coding RNA in 8q24 with prostate cancer susceptibility. Cancer Sci. 2011, 102, 245–252. [Google Scholar] [CrossRef]
  161. Ren, S.; Wang, F.; Shen, J.; Sun, Y.; Xu, W.; Lu, J.; Wei, M.; Xu, C.; Wu, C.; Zhang, Z.; et al. Long non-coding RNA metastasis associated in lung adenocarcinoma transcript 1 derived miniRNA as a novel plasma-based biomarker for diagnosing prostate cancer. Eur. J. Cancer 2013, 49, 2949–2959. [Google Scholar] [CrossRef]
  162. Prensner, J.R.; Chen, W.; Han, S.; Iyer, M.K.; Cao, Q.; Kothari, V.; Evans, J.R.; Knudsen, K.E.; Paulsen, M.T.; Ljungman, M.; et al. The long non-coding RNA PCAT-1 promotes prostate cancer cell proliferation through cMyc. Neoplasia 2014, 16, 900–908. [Google Scholar] [CrossRef] [PubMed]
  163. Yaman Agaoglu, F.; Kovancilar, M.; Dizdar, Y.; Darendeliler, E.; Holdenrieder, S.; Dalay, N.; Gezer, U. Investigation of miR-21, miR-141, and miR-221 in blood circulation of patients with prostate cancer. Tumour Biol. 2011, 32, 583–588. [Google Scholar] [CrossRef] [PubMed]
  164. Brase, J.C.; Johannes, M.; Schlomm, T.; Fälth, M.; Haese, A.; Steuber, T.; Beissbarth, T.; Kuner, R.; Sültmann, H. Circulating miRNAs are correlated with tumor progression in prostate cancer. Int. J. Cancer 2011, 128, 608–616. [Google Scholar] [CrossRef] [PubMed]
  165. Shi, T.; Gao, G.; Cao, Y. Long Noncoding RNAs as Novel Biomarkers Have a Promising Future in Cancer Diagnostics. Dis. Markers 2016, 2016, 9085195. [Google Scholar] [CrossRef]
  166. Xu, M.D.; Qi, P.; Weng, W.W.; Shen, X.H.; Ni, S.J.; Dong, L.; Huang, D.; Tan, C.; Sheng, W.Q.; Zhou, X.Y.; et al. Long non-coding RNA LSINCT5 predicts negative prognosis and exhibits oncogenic activity in gastric cancer. Medicine 2014, 93, e303. [Google Scholar] [CrossRef]
  167. Zhou, X.; Yin, C.; Dang, Y.; Ye, F.; Zhang, G. Identification of the long non-coding RNA H19 in plasma as a novel biomarker for diagnosis of gastric cancer. Sci. Rep. 2015, 5, 11516. [Google Scholar] [CrossRef]
  168. Kong, R.; Zhang, E.B.; Yin, D.D.; You, L.H.; Xu, T.P.; Chen, W.M.; Xia, R.; Wan, L.; Sun, M.; Wang, Z.X.; et al. Long noncoding RNA PVT1 indicates a poor prognosis of gastric cancer and promotes cell proliferation through epigenetically regulating p15 and p16. Mol. Cancer 2015, 14, 82. [Google Scholar] [CrossRef]
  169. Liu, R.; Zhang, C.; Hu, Z.; Li, G.; Wang, C.; Yang, C.; Huang, D.; Chen, X.; Zhang, H.; Zhuang, R.; et al. A five-microRNA signature identified from genome-wide serum microRNA expression profiling serves as a fingerprint for gastric cancer diagnosis. Eur. J. Cancer 2011, 47, 784–791. [Google Scholar] [CrossRef]
  170. Tsujiura, M.; Ichikawa, D.; Komatsu, S.; Shiozaki, A.; Takeshita, H.; Kosuga, T.; Konishi, H.; Morimura, R.; Deguchi, K.; Fujiwara, H.; et al. Circulating microRNAs in plasma of patients with gastric cancers. Br. J. Cancer 2010, 102, 1174–1179. [Google Scholar] [CrossRef]
  171. Peng, W.; Gao, W.; Feng, J. Long noncoding RNA HULC is a novel biomarker of poor prognosis in patients with pancreatic cancer. Med. Oncol. 2014, 31, 346. [Google Scholar] [CrossRef]
  172. Kim, K.; Jutooru, I.; Chadalapaka, G.; Johnson, G.; Frank, J.; Burghardt, R.; Kim, S.; Safe, S. HOTAIR is a negative prognostic factor and exhibits pro-oncogenic activity in pancreatic cancer. Oncogene 2013, 32, 1616–1625. [Google Scholar] [CrossRef] [PubMed]
  173. Wang, J.; Chen, J.; Chang, P.; LeBlanc, A.; Li, D.; Abbruzzesse, J.L.; Frazier, M.L.; Killary, A.M.; Sen, S. MicroRNAs in plasma of pancreatic ductal adenocarcinoma patients as novel blood-based biomarkers of disease. Cancer Prev. Res. 2009, 2, 807–813. [Google Scholar] [CrossRef] [PubMed]
  174. Miller, T.E.; Ghoshal, K.; Ramaswamy, B.; Roy, S.; Datta, J.; Shapiro, C.L.; Jacob, S.; Majumder, S. MicroRNA-221/222 confers tamoxifen resistance in breast cancer by targeting p27Kip1. J. Biol. Chem. 2008, 283, 29897–29903. [Google Scholar] [CrossRef] [PubMed]
  175. Li, A.; Omura, N.; Hong, S.M.; Vincent, A.; Walter, K.; Griffith, M.; Borges, M.; Goggins, M. Pancreatic cancers epigenetically silence SIP1 and hypomethylate and overexpress miR-200a/200b in association with elevated circulating miR-200a and miR-200b levels. Cancer Res. 2010, 70, 5226–5237. [Google Scholar] [CrossRef] [PubMed]
  176. Weber, D.G.; Johnen, G.; Casjens, S.; Bryk, O.; Pesch, B.; Jöckel, K.H.; Kollmeier, J.; Brüning, T. Evaluation of long noncoding RNA MALAT1 as a candidate blood-based biomarker for the diagnosis of non-small cell lung cancer. BMC Res. Notes 2013, 6, 518. [Google Scholar] [CrossRef]
  177. Zeng, Z.; Bo, H.; Gong, Z.; Lian, Y.; Li, X.; Li, X.; Zhang, W.; Deng, H.; Zhou, M.; Peng, S.; et al. AFAP1-AS1, a long noncoding RNA upregulated in lung cancer and promotes invasion and metastasis. Tumour Biol. 2016, 37, 729–737. [Google Scholar] [CrossRef]
  178. Liu, X.H.; Liu, Z.L.; Sun, M.; Liu, J.; Wang, Z.X.; De, W. The long non-coding RNA HOTAIR indicates a poor prognosis and promotes metastasis in non-small cell lung cancer. BMC Cancer 2013, 13, 464. [Google Scholar] [CrossRef]
  179. Qiu, M.; Xu, Y.; Yang, X.; Wang, J.; Hu, J.; Xu, L.; Yin, R. CCAT2 is a lung adenocarcinoma-specific long non-coding RNA and promotes invasion of non-small cell lung cancer. Tumour Biol. 2014, 35, 5375–5380. [Google Scholar] [CrossRef]
  180. Nie, F.Q.; Zhu, Q.; Xu, T.P.; Zou, Y.F.; Xie, M.; Sun, M.; Xia, R.; Lu, K.H. Long non-coding RNA MVIH indicates a poor prognosis for non-small cell lung cancer and promotes cell proliferation and invasion. Tumour Biol. 2014, 35, 7587–7594. [Google Scholar] [CrossRef]
  181. White, N.M.; Cabanski, C.R.; Silva-Fisher, J.M.; Dang, H.X.; Govindan, R.; Maher, C.A. Transcriptome sequencing reveals altered long intergenic non-coding RNAs in lung cancer. Genome Biol. 2014, 15, 429. [Google Scholar] [CrossRef]
  182. Qiu, M.; Xu, Y.; Wang, J.; Zhang, E.; Sun, M.; Zheng, Y.; Li, M.; Xia, W.; Feng, D.; Yin, R.; et al. A novel lncRNA, LUADT1, promotes lung adenocarcinoma proliferation via the epigenetic suppression of p27. Cell Death Dis. 2015, 6, e1858. [Google Scholar] [CrossRef]
  183. Silva, J.; García, V.; Zaballos, Á.; Provencio, M.; Lombardía, L.; Almonacid, L.; García, J.M.; Domínguez, G.; Peña, C.; Diaz, R.; et al. Vesicle-related microRNAs in plasma of nonsmall cell lung cancer patients and correlation with survival. Eur. Respir. J. 2011, 37, 617–623. [Google Scholar] [CrossRef] [PubMed]
  184. Hu, Z.; Chen, X.; Zhao, Y.; Tian, T.; Jin, G.; Shu, Y.; Chen, Y.; Xu, L.; Zen, K.; Zhang, C.; et al. Serum microRNA signatures identified in a genome-wide serum microRNA expression profiling predict survival of non-small-cell lung cancer. J. Clin. Oncol. 2010, 28, 1721–1726. [Google Scholar] [CrossRef] [PubMed]
  185. Heegaard, N.H.; Schetter, A.J.; Welsh, J.A.; Yoneda, M.; Bowman, E.D.; Harris, C.C. Circulating micro-RNA expression profiles in early stage nonsmall cell lung cancer. Int. J. Cancer 2012, 130, 1378–1386. [Google Scholar] [CrossRef] [PubMed]
  186. Wei, J.; Gao, W.; Zhu, C.J.; Liu, Y.Q.; Mei, Z.; Cheng, T.; Shu, Y.Q. Identification of plasma microRNA-21 as a biomarker for early detection and chemosensitivity of non-small cell lung cancer. Chin. J. Cancer 2011, 30, 407–414. [Google Scholar] [CrossRef] [PubMed]
  187. Chen, X.; Ba, Y.; Ma, L.; Cai, X.; Yin, Y.; Wang, K.; Guo, J.; Zhang, Y.; Chen, J.; Guo, X.; et al. Characterization of microRNAs in serum: A novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res. 2008, 18, 997–1006. [Google Scholar] [CrossRef]
  188. Zhou, C.C.; Yang, F.; Yuan, S.X.; Ma, J.Z.; Liu, F.; Yuan, J.H.; Bi, F.R.; Lin, K.Y.; Yin, J.H.; Cao, G.W.; et al. Systemic genome screening identifies the outcome associated focal loss of long noncoding RNA PRAL in hepatocellular carcinoma. Hepatology 2016, 63, 850–863. [Google Scholar] [CrossRef]
  189. Malakar, P.; Shilo, A.; Mogilevsky, A.; Stein, I.; Pikarsky, E.; Nevo, Y.; Benyamini, H.; Elgavish, S.; Zong, X.; Prasanth, K.V.; et al. Long Noncoding RNA MALAT1 Promotes Hepatocellular Carcinoma Development by SRSF1 Upregulation and mTOR Activation. Cancer Res. 2017, 77, 1155–1167. [Google Scholar] [CrossRef]
  190. Geng, Y.J.; Xie, S.L.; Li, Q.; Ma, J.; Wang, G.Y. Large intervening non-coding RNA HOTAIR is associated with hepatocellular carcinoma progression. J. Int. Med. Res. 2011, 39, 2119–2128. [Google Scholar] [CrossRef]
  191. Tang, J.; Jiang, R.; Deng, L.; Zhang, X.; Wang, K.; Sun, B. Circulation long non-coding RNAs act as biomarkers for predicting tumorigenesis and metastasis in hepatocellular carcinoma. Oncotarget 2015, 6, 4505–4515. [Google Scholar] [CrossRef]
  192. Zhou, T.; Gao, Y. Increased expression of LncRNA BANCR and its prognostic significance in human hepatocellular carcinoma. World J. Surg. Oncol. 2016, 14, 8. [Google Scholar] [CrossRef] [PubMed]
  193. Zhang, T.; Cao, C.; Wu, D.; Liu, L. SNHG3 correlates with malignant status and poor prognosis in hepatocellular carcinoma. Tumour Biol. 2016, 37, 2379–2385. [Google Scholar] [CrossRef] [PubMed]
  194. Shi, Y.; Song, Q.; Yu, S.; Hu, D.; Zhuang, X. Microvascular invasion in hepatocellular carcinoma overexpression promotes cell proliferation and inhibits cell apoptosis of hepatocellular carcinoma via inhibiting miR-199a expression. Onco Targets Ther. 2015, 8, 2303–2310. [Google Scholar] [CrossRef] [PubMed]
  195. Huang, M.D.; Chen, W.M.; Qi, F.Z.; Xia, R.; Sun, M.; Xu, T.P.; Yin, L.; Zhang, E.B.; De, W.; Shu, Y.Q. Long non-coding RNA ANRIL is upregulated in hepatocellular carcinoma and regulates cell apoptosis by epigenetic silencing of KLF2. J. Hematol. Oncol. 2015, 8, 50. [Google Scholar] [CrossRef] [PubMed]
  196. Panzitt, K.; Tschernatsch, M.M.; Guelly, C.; Moustafa, T.; Stradner, M.; Strohmaier, H.M.; Buck, C.R.; Denk, H.; Schroeder, R.; Trauner, M.; et al. Characterization of HULC, a novel gene with striking up-regulation in hepatocellular carcinoma, as noncoding RNA. Gastroenterology 2007, 132, 330–342. [Google Scholar] [CrossRef]
  197. Shigoka, M.; Tsuchida, A.; Matsudo, T.; Nagakawa, Y.; Saito, H.; Suzuki, Y.; Aoki, T.; Murakami, Y.; Toyoda, H.; Kumada, T.; et al. Deregulation of miR-92a expression is implicated in hepatocellular carcinoma development. Pathol. Int. 2010, 60, 351–357. [Google Scholar] [CrossRef]
  198. Xu, J.; Wu, C.; Che, X.; Wang, L.; Yu, D.; Zhang, T.; Huang, L.; Li, H.; Tan, W.; Wang, C.; et al. Circulating microRNAs, miR-21, miR-122, and miR-223, in patients with hepatocellular carcinoma or chronic hepatitis. Mol. Carcinog. 2011, 50, 136–142. [Google Scholar] [CrossRef]
  199. Yamamoto, Y.; Kosaka, N.; Tanaka, M.; Koizumi, F.; Kanai, Y.; Mizutani, T.; Murakami, Y.; Kuroda, M.; Miyajima, A.; Kato, T.; et al. MicroRNA-500 as a potential diagnostic marker for hepatocellular carcinoma. Biomarkers 2009, 14, 529–538. [Google Scholar] [CrossRef]
  200. Gui, J.; Tian, Y.; Wen, X.; Zhang, W.; Zhang, P.; Gao, J.; Run, W.; Tian, L.; Jia, X.; Gao, Y. Serum microRNA characterization identifies miR-885-5p as a potential marker for detecting liver pathologies. Clin. Sci. 2011, 120, 183–193. [Google Scholar] [CrossRef]
  201. Tang, H.; Wu, Z.; Zhang, J.; Su, B. Salivary lncRNA as a potential marker for oral squamous cell carcinoma diagnosis. Mol. Med. Rep. 2013, 7, 761–766. [Google Scholar] [CrossRef]
  202. Yang, Y.T.; Wang, Y.F.; Lai, J.Y.; Shen, S.Y.; Wang, F.; Kong, J.; Zhang, W.; Yang, H.Y. Long non-coding RNA UCA1 contributes to the progression of oral squamous cell carcinoma by regulating the WNT/β-catenin signaling pathway. Cancer Sci. 2016, 107, 1581–1589. [Google Scholar] [CrossRef]
  203. Wu, Y.; Zhang, L.; Zhang, L.; Wang, Y.; Li, H.; Ren, X.; Wei, F.; Yu, W.; Liu, T.; Wang, X.; et al. Long non-coding RNA HOTAIR promotes tumor cell invasion and metastasis by recruiting EZH2 and repressing E-cadherin in oral squamous cell carcinoma. Int. J. Oncol. 2015, 46, 2586–2594. [Google Scholar] [CrossRef] [PubMed]
  204. Lin, S.C.; Liu, C.J.; Lin, J.A.; Chiang, W.F.; Hung, P.S.; Chang, K.W. miR-24 up-regulation in oral carcinoma: Positive association from clinical and in vitro analysis. Oral. Oncol. 2010, 46, 204–208. [Google Scholar] [CrossRef] [PubMed]
  205. Liu, C.J.; Kao, S.Y.; Tu, H.F.; Tsai, M.M.; Chang, K.W.; Lin, S.C. Increase of microRNA miR-31 level in plasma could be a potential marker of oral cancer. Oral. Dis. 2010, 16, 360–364. [Google Scholar] [CrossRef] [PubMed]
  206. Dallosso, A.R.; Hancock, A.L.; Malik, S.; Salpekar, A.; King-Underwood, L.; Pritchard-Jones, K.; Peters, J.; Moorwood, K.; Ward, A.; Malik, K.T.; et al. Alternately spliced WT1 antisense transcripts interact with WT1 sense RNA and show epigenetic and splicing defects in cancer. RNA 2007, 13, 2287–2299. [Google Scholar] [CrossRef] [PubMed]
  207. Tanaka, M.; Oikawa, K.; Takanashi, M.; Kudo, M.; Ohyashiki, J.; Ohyashiki, K.; Kuroda, M. Down-regulation of miR-92 in human plasma is a novel marker for acute leukemia patients. PLoS ONE 2009, 4, e5532. [Google Scholar] [CrossRef]
  208. Yang, M.; Zhai, X.; Xia, B.; Wang, Y.; Lou, G. Long noncoding RNA CCHE1 promotes cervical cancer cell proliferation via upregulating PCNA. Tumour Biol. 2015, 36, 7615–7622. [Google Scholar] [CrossRef]
  209. Lee, M.; Kim, H.J.; Kim, S.W.; Park, S.A.; Chun, K.H.; Cho, N.H.; Song, Y.S.; Kim, Y.T. The long non-coding RNA HOTAIR increases tumour growth and invasion in cervical cancer by targeting the Notch pathway. Oncotarget 2016, 7, 44558–44571. [Google Scholar] [CrossRef]
  210. Wu, L.; Jin, L.; Zhang, W.; Zhang, L. Roles of Long Non-Coding RNA CCAT2 in Cervical Cancer Cell Growth and Apoptosis. Med. Sci. Monit. 2016, 22, 875–879. [Google Scholar] [CrossRef]
  211. Causin, R.L.; Freitas, A.J.A.; Hidalgo Filho, C.M.T.; Reis, R.D.; Reis, R.M.; Marques, M.M.C. A Systematic Review of MicroRNAs Involved in Cervical Cancer Progression. Cells 2021, 10, 668. [Google Scholar] [CrossRef]
  212. Lessard, L.; Liu, M.; Marzese, D.M.; Wang, H.; Chong, K.; Kawas, N.; Donovan, N.C.; Kiyohara, E.; Hsu, S.; Nelson, N.; et al. The CASC15 Long Intergenic Noncoding RNA Locus Is Involved in Melanoma Progression and Phenotype Switching. J. Investig. Dermatol. 2015, 135, 2464–2474. [Google Scholar] [CrossRef] [PubMed]
  213. Khaitan, D.; Dinger, M.E.; Mazar, J.; Crawford, J.; Smith, M.A.; Mattick, J.S.; Perera, R.J. The melanoma-upregulated long noncoding RNA SPRY4-IT1 modulates apoptosis and invasion. Cancer Res. 2011, 71, 3852–3862. [Google Scholar] [CrossRef] [PubMed]
  214. Mazar, J.; Zhao, W.; Khalil, A.M.; Lee, B.; Shelley, J.; Govindarajan, S.S.; Yamamoto, F.; Ratnam, M.; Aftab, M.N.; Collins, S.; et al. The functional characterization of long noncoding RNA SPRY4-IT1 in human melanoma cells. Oncotarget 2014, 5, 8959–8969. [Google Scholar] [CrossRef]
  215. Luo, M.; Li, Z.; Wang, W.; Zeng, Y.; Liu, Z.; Qiu, J. Long non-coding RNA H19 increases bladder cancer metastasis by associating with EZH2 and inhibiting E-cadherin expression. Cancer Lett. 2013, 333, 213–221. [Google Scholar] [CrossRef]
  216. Srivastava, A.K.; Singh, P.K.; Rath, S.K.; Dalela, D.; Goel, M.M.; Bhatt, M.L. Appraisal of diagnostic ability of UCA1 as a biomarker of carcinoma of the urinary bladder. Tumour Biol. 2014, 35, 11435–11442. [Google Scholar] [CrossRef]
  217. Homami, A.; Ghazi, F. MicroRNAs as biomarkers associated with bladder cancer. Med. J. Islam. Repub. Iran. 2016, 30, 475. [Google Scholar]
  218. Ziv, E.; Durack, J.C.; Solomon, S.B. The Importance of Biopsy in the Era of Molecular Medicine. Cancer J. 2016, 22, 418–422. [Google Scholar] [CrossRef]
  219. Hirahata, T.; Ul Quraish, R.; Quraish, A.U.; Ul Quraish, S.; Naz, M.; Razzaq, M.A. Liquid Biopsy: A Distinctive Approach to the Diagnosis and Prognosis of Cancer. Cancer Inform. 2022, 21, 11769351221076062. [Google Scholar] [CrossRef]
  220. Connal, S.; Cameron, J.M.; Sala, A.; Brennan, P.M.; Palmer, D.S.; Palmer, J.D.; Perlow, H.; Baker, M.J. Liquid biopsies: The future of cancer early detection. J. Transl. Med. 2023, 21, 118. [Google Scholar] [CrossRef]
  221. Eslami, S.Z.; Cortes-Hernandez, L.E.; Cayrefourcq, L.; Alix-Panabieres, C. The Different Facets of Liquid Biopsy: A Kaleidoscopic View. Cold Spring Harb. Perspect. Med. 2020, 10, a037333. [Google Scholar] [CrossRef]
  222. Wang, X.; Wang, L.; Lin, H.; Zhu, Y.; Huang, D.; Lai, M.; Xi, X.; Huang, J.; Zhang, W.; Zhong, T. Research progress of CTC, ctDNA, and EVs in cancer liquid biopsy. Front. Oncol. 2024, 14, 1303335. [Google Scholar] [CrossRef] [PubMed]
  223. Mobadersany, P.; Yousefi, S.; Amgad, M.; Gutman, D.A.; Barnholtz-Sloan, J.S.; Velázquez Vega, J.E.; Brat, D.J.; Cooper, L.A.D. Predicting cancer outcomes from histology and genomics using convolutional networks. Proc. Natl. Acad. Sci. USA 2018, 115, E2970–E2979. [Google Scholar] [CrossRef] [PubMed]
  224. Testa, U.; Castelli, G.; Pelosi, E. Cellular and Molecular Mechanisms Underlying Prostate Cancer Development: Therapeutic Implications. Medicines 2019, 6, 82. [Google Scholar] [CrossRef]
  225. Ahmed, A.A.; Abedalthagafi, M. Cancer diagnostics: The journey from histomorphology to molecular profiling. Oncotarget 2016, 7, 58696–58708. [Google Scholar] [CrossRef] [PubMed]
  226. Yoshimura, A.; Imoto, I.; Iwata, H. Functions of Breast Cancer Predisposition Genes: Implications for Clinical Management. Int. J. Mol. Sci. 2022, 23, 7481. [Google Scholar] [CrossRef]
  227. Hegde, M.; Blazo, M.; Chong, B.; Prior, T.; Richards, C. Assay validation for identification of hereditary nonpolyposis colon cancer-causing mutations in mismatch repair genes MLH1, MSH2, and MSH6. J. Mol. Diagn. 2005, 7, 525–534. [Google Scholar] [CrossRef]
  228. Cruz-Gálvez, C.C.; Ordaz-Favila, J.C.; Villar-Calvo, V.M.; Cancino-Marentes, M.E.; Bosch-Canto, V. Retinoblastoma: Review and new insights. Front. Oncol. 2022, 12, 963780. [Google Scholar] [CrossRef]
  229. Li, S.; Silvestri, V.; Leslie, G.; Rebbeck, T.R.; Neuhausen, S.L.; Hopper, J.L.; Nielsen, H.R.; Lee, A.; Yang, X.; McGuffog, L.; et al. Cancer Risks Associated With BRCA1 and BRCA2 Pathogenic Variants. J. Clin. Oncol. 2022, 40, 1529–1541. [Google Scholar] [CrossRef]
  230. Chen, X.; Zhang, T.; Su, W.; Dou, Z.; Zhao, D.; Jin, X.; Lei, H.; Wang, J.; Xie, X.; Cheng, B.; et al. Mutant p53 in cancer: From molecular mechanism to therapeutic modulation. Cell Death Dis. 2022, 13, 974. [Google Scholar] [CrossRef]
  231. Cummings, S.; Alfonso, A.; Hughes, E.; Kucera, M.; Mabey, B.; Singh, N.; Eng, C. Cancer Risk Associated With PTEN Pathogenic Variants Identified Using Multigene Hereditary Cancer Panel Testing. JCO Precis. Oncol. 2023, 7, e2200415. [Google Scholar] [CrossRef]
  232. Vogt, S.; Jones, N.; Christian, D.; Engel, C.; Nielsen, M.; Kaufmann, A.; Steinke, V.; Vasen, H.F.; Propping, P.; Sampson, J.R.; et al. Expanded extracolonic tumor spectrum in MUTYH-associated polyposis. Gastroenterology 2009, 137, 1976–1985.e10. [Google Scholar] [CrossRef] [PubMed]
  233. Korsse, S.E.; Peppelenbosch, M.P.; van Veelen, W. Targeting LKB1 signaling in cancer. Biochim. Biophys. Acta 2013, 1835, 194–210. [Google Scholar] [CrossRef] [PubMed]
  234. Crosbie, E.J.; Ryan, N.A.J.; McVey, R.J.; Lalloo, F.; Bowers, N.; Green, K.; Woodward, E.R.; Clancy, T.; Bolton, J.; Wallace, A.J.; et al. Assessment of mismatch repair deficiency in ovarian cancer. J. Med. Genet. 2021, 58, 687–691. [Google Scholar] [CrossRef] [PubMed]
  235. Castro, E.; Eeles, R. The role of BRCA1 and BRCA2 in prostate cancer. Asian J. Androl. 2012, 14, 409–414. [Google Scholar] [CrossRef]
  236. Sharma, M.; Yang, Z.; Miyamoto, H. Loss of DNA mismatch repair proteins in prostate cancer. Medicine 2020, 99, e20124. [Google Scholar] [CrossRef]
  237. Devico Marciano, N.; Kroening, G.; Dayyani, F.; Zell, J.A.; Lee, F.C.; Cho, M.; Valerin, J.G. BRCA-Mutated Pancreatic Cancer: From Discovery to Novel Treatment Paradigms. Cancers 2022, 14, 2453. [Google Scholar] [CrossRef]
  238. Klatte, D.C.F.; Wallace, M.B.; Löhr, M.; Bruno, M.J.; van Leerdam, M.E. Hereditary pancreatic cancer. Best. Pract. Res. Clin. Gastroenterol. 2022, 58–59, 101783. [Google Scholar] [CrossRef]
  239. Corbo, V.; Dalai, I.; Scardoni, M.; Barbi, S.; Beghelli, S.; Bersani, S.; Albarello, L.; Doglioni, C.; Schott, C.; Capelli, P.; et al. MEN1 in pancreatic endocrine tumors: Analysis of gene and protein status in 169 sporadic neoplasms reveals alterations in the vast majority of cases. Endocr. Relat. Cancer 2010, 17, 771–783. [Google Scholar] [CrossRef]
  240. Buckley, K.H.; Niccum, B.A.; Maxwell, K.N.; Katona, B.W. Gastric Cancer Risk and Pathogenesis in BRCA1 and BRCA2 Carriers. Cancers 2022, 14, 5953. [Google Scholar] [CrossRef]
  241. Salari, S.; Ghadyani, M.; Karimi, M.; Mortezazadeh, M.; Vahedifard, F. Immunohistochemical Expression Pattern of MLH1, MSH2, MSH6, and PMS2 in Tumor Specimen of Iranian Gastric Carcinoma Patients. J. Gastrointest. Cancer 2022, 53, 192–196. [Google Scholar] [CrossRef]
  242. Fang, D.C.; Luo, Y.H.; Yang, S.M.; Li, X.A.; Ling, X.L.; Fang, L. Mutation analysis of APC gene in gastric cancer with microsatellite instability. World J. Gastroenterol. 2002, 8, 787–791. [Google Scholar] [CrossRef] [PubMed]
  243. Jiang, S.; Chen, R.; Yu, J.; Li, N.; Ke, R.; Luo, L.; Zou, J.; Zhang, J.; Zhang, K.; Lu, N.; et al. Clinical significance and role of LKB1 in gastric cancer. Mol. Med. Rep. 2016, 13, 249–256. [Google Scholar] [CrossRef] [PubMed]
  244. Sok, C.; Ajay, P.S.; Tsagkalidis, V.; Kooby, D.A.; Shah, M.M. Management of Gastric Neuroendocrine Tumors: A Review. Ann. Surg. Oncol. 2024, 31, 1509–1518. [Google Scholar] [CrossRef]
  245. Karahan, B.; Argon, A.; Yıldırım, M.; Vardar, E. Relationship between MLH-1, MSH-2, PMS-2,MSH-6 expression and clinicopathological features in colorectal cancer. Int. J. Clin. Exp. Pathol. 2015, 8, 4044–4053. [Google Scholar]
  246. van der Post, R.S.; Kiemeney, L.A.; Ligtenberg, M.J.; Witjes, J.A.; Hulsbergen-van de Kaa, C.A.; Bodmer, D.; Schaap, L.; Kets, C.M.; van Krieken, J.H.; Hoogerbrugge, N. Risk of urothelial bladder cancer in Lynch syndrome is increased, in particular among MSH2 mutation carriers. J. Med. Genet. 2010, 47, 464–470. [Google Scholar] [CrossRef]
  247. Abdel-Wahab, R.; Yap, T.A.; Madison, R.; Pant, S.; Cooke, M.; Wang, K.; Zhao, H.; Bekaii-Saab, T.; Karatas, E.; Kwong, L.N.; et al. Genomic profiling reveals high frequency of DNA repair genetic aberrations in gallbladder cancer. Sci. Rep. 2020, 10, 22087. [Google Scholar] [CrossRef]
  248. Berg, H.F.; Engerud, H.; Myrvold, M.; Lien, H.E.; Hjelmeland, M.E.; Halle, M.K.; Woie, K.; Hoivik, E.A.; Haldorsen, I.S.; Vintermyr, O.; et al. Mismatch repair markers in preoperative and operative endometrial cancer samples; expression concordance and prognostic value. Br. J. Cancer 2023, 128, 647–655. [Google Scholar] [CrossRef]
  249. Stavropoulos, A.; Varras, M.; Vasilakaki, T.; Varra, V.K.; Tsavari, A.; Varra, F.N.; Nonni, A.; Kavantzas, N.; Lazaris, A.C. Expression of p53 and PTEN in human primary endometrial carcinomas: Clinicopathological and immunohistochemical analysis and study of their concomitant expression. Oncol. Lett. 2019, 17, 4575–4589. [Google Scholar] [CrossRef]
  250. Lintas, C.; Canalis, B.; Azzarà, A.; Sabarese, G.; Perrone, G.; Gurrieri, F. Exploring the Role of the MUTYH Gene in Breast, Ovarian and Endometrial Cancer. Genes 2024, 15, 554. [Google Scholar] [CrossRef]
  251. Thoenen, E.; Curl, A.; Iwakuma, T. TP53 in bone and soft tissue sarcomas. Pharmacol. Ther. 2019, 202, 149–164. [Google Scholar] [CrossRef]
  252. Santini, V.; Stahl, M.; Sallman, D.A. TP53 Mutations in Acute Leukemias and Myelodysplastic Syndromes: Insights and Treatment Updates. Am. Soc. Clin. Oncol. Educ. Book. 2024, 44, e432650. [Google Scholar] [CrossRef] [PubMed]
  253. Aguissa-Touré, A.H.; Li, G. Genetic alterations of PTEN in human melanoma. Cell Mol. Life Sci. 2012, 69, 1475–1491. [Google Scholar] [CrossRef]
  254. Su, L.K.; Abdalla, E.K.; Law, C.H.; Kohlmann, W.; Rashid, A.; Vauthey, J.N. Biallelic inactivation of the APC gene is associated with hepatocellular carcinoma in familial adenomatous polyposis coli. Cancer 2001, 92, 332–339. [Google Scholar] [CrossRef]
  255. Yun, J.; Li, Y.; Xu, C.T.; Pan, B.R. Epidemiology and Rb1 gene of retinoblastoma. Int. J. Ophthalmol. 2011, 4, 103–109. [Google Scholar] [CrossRef]
  256. Unger, M.; Kather, J.N. A systematic analysis of deep learning in genomics and histopathology for precision oncology. BMC Med. Genomics 2024, 17, 48. [Google Scholar] [CrossRef]
  257. Unger, M.; Kather, J.N. Deep learning in cancer genomics and histopathology. Genome Med. 2024, 16, 44. [Google Scholar] [CrossRef]
  258. Nafees, A.; Khan, M.; Chow, R.; Fazelzad, R.; Hope, A.; Liu, G.; Letourneau, D.; Raman, S. Evaluation of clinical decision support systems in oncology: An updated systematic review. Crit. Rev. Oncol. Hematol. 2023, 192, 104143. [Google Scholar] [CrossRef]
  259. Restrepo, J.C.; Duenas, D.; Corredor, Z.; Liscano, Y. Advances in Genomic Data and Biomarkers: Revolutionizing NSCLC Diagnosis and Treatment. Cancers 2023, 15, 3474. [Google Scholar] [CrossRef]
  260. Schwalbe, N.; Wahl, B.; Song, J.; Lehtimaki, S. Data Sharing and Global Public Health: Defining What We Mean by Data. Front. Digit. Health 2020, 2, 612339. [Google Scholar] [CrossRef]
  261. Li, A.; Bergan, R.C. Clinical trial design: Past, present, and future in the context of big data and precision medicine. Cancer 2020, 126, 4838–4846. [Google Scholar] [CrossRef]
  262. Yang, X.; Huang, K.; Yang, D.; Zhao, W.; Zhou, X. Biomedical Big Data Technologies, Applications, and Challenges for Precision Medicine: A Review. Glob. Chall. 2024, 8, 2300163. [Google Scholar] [CrossRef]
  263. Alowais, S.A.; Alghamdi, S.S.; Alsuhebany, N.; Alqahtani, T.; Alshaya, A.I.; Almohareb, S.N.; Aldairem, A.; Alrashed, M.; Bin Saleh, K.; Badreldin, H.A.; et al. Revolutionizing healthcare: The role of artificial intelligence in clinical practice. BMC Med. Educ. 2023, 23, 689. [Google Scholar] [CrossRef]
  264. Badr, Y.; Abdul Kader, L.; Shamayleh, A. The Use of Big Data in Personalized Healthcare to Reduce Inventory Waste and Optimize Patient Treatment. J. Pers. Med. 2024, 14, 383. [Google Scholar] [CrossRef]
  265. Changhez, J.; James, S.; Jamala, F.; Khan, S.; Khan, M.Z.; Gul, S.; Zainab, I. Evaluating the Efficacy and Accuracy of AI-Assisted Diagnostic Techniques in Endometrial Carcinoma: A Systematic Review. Cureus 2024, 16, e60973. [Google Scholar] [CrossRef]
  266. Perivolaris, A.; Adams-McGavin, C.; Madan, Y.; Kishibe, T.; Antoniou, T.; Mamdani, M.; Jung, J.J. Quality of interaction between clinicians and artificial intelligence systems. A systematic review. Future Healthc. J. 2024, 11, 100172. [Google Scholar] [CrossRef]
  267. Norori, N.; Hu, Q.; Aellen, F.M.; Faraci, F.D.; Tzovara, A. Addressing bias in big data and AI for health care: A call for open science. Patterns 2021, 2, 100347. [Google Scholar] [CrossRef]
  268. Arora, A.; Alderman, J.E.; Palmer, J.; Ganapathi, S.; Laws, E.; McCradden, M.D.; Oakden-Rayner, L.; Pfohl, S.R.; Ghassemi, M.; McKay, F.; et al. The value of standards for health datasets in artificial intelligence-based applications. Nat. Med. 2023, 29, 2929–2938. [Google Scholar] [CrossRef]
  269. Khalid, N.; Qayyum, A.; Bilal, M.; Al-Fuqaha, A.; Qadir, J. Privacy-preserving artificial intelligence in healthcare: Techniques and applications. Comput. Biol. Med. 2023, 158, 106848. [Google Scholar] [CrossRef]
  270. Khosravi, M.; Zare, Z.; Mojtabaeian, S.M.; Izadi, R. Artificial Intelligence and Decision-Making in Healthcare: A Thematic Analysis of a Systematic Review of Reviews. Health Serv. Res. Manag. Epidemiol. 2024, 11, 23333928241234863. [Google Scholar] [CrossRef]
  271. Naik, N.; Hameed, B.M.Z.; Shetty, D.K.; Swain, D.; Shah, M.; Paul, R.; Aggarwal, K.; Ibrahim, S.; Patil, V.; Smriti, K.; et al. Legal and Ethical Consideration in Artificial Intelligence in Healthcare: Who Takes Responsibility? Front. Surg. 2022, 9, 862322. [Google Scholar] [CrossRef]
  272. Farhud, D.D.; Zokaei, S. Ethical Issues of Artificial Intelligence in Medicine and Healthcare. Iran. J. Public. Health 2021, 50, i–v. [Google Scholar] [CrossRef]
  273. Saeed, R.F.; Awan, U.A.; Saeed, S.; Mumtaz, S.; Akhtar, N.; Aslam, S. Targeted Therapy and Personalized Medicine. Cancer Treat. Res. 2023, 185, 177–205. [Google Scholar] [CrossRef]
  274. Goetz, L.H.; Schork, N.J. Personalized medicine: Motivation, challenges, and progress. Fertil. Steril. 2018, 109, 952–963. [Google Scholar] [CrossRef]
  275. Wang, R.C.; Wang, Z. Precision Medicine: Disease Subtyping and Tailored Treatment. Cancers 2023, 15, 3837. [Google Scholar] [CrossRef]
  276. Hoeben, A.; Joosten, E.A.J.; van den Beuken-van Everdingen, M.H.J. Personalized Medicine: Recent Progress in Cancer Therapy. Cancers 2021, 13, 242. [Google Scholar] [CrossRef]
  277. Krzyszczyk, P.; Acevedo, A.; Davidoff, E.J.; Timmins, L.M.; Marrero-Berrios, I.; Patel, M.; White, C.; Lowe, C.; Sherba, J.J.; Hartmanshenn, C.; et al. The growing role of precision and personalized medicine for cancer treatment. Technology 2018, 6, 79–100. [Google Scholar] [CrossRef]
  278. Rulten, S.L.; Grose, R.P.; Gatz, S.A.; Jones, J.L.; Cameron, A.J.M. The Future of Precision Oncology. Int. J. Mol. Sci. 2023, 24, 12613. [Google Scholar] [CrossRef]
  279. Yadav, D.; Patil-Takbhate, B.; Khandagale, A.; Bhawalkar, J.; Tripathy, S.; Khopkar-Kale, P. Next-Generation sequencing transforming clinical practice and precision medicine. Clin. Chim. Acta 2023, 551, 117568. [Google Scholar] [CrossRef]
  280. Colomer, R.; Miranda, J.; Romero-Laorden, N.; Hornedo, J.; Gonzalez-Cortijo, L.; Mouron, S.; Bueno, M.J.; Mondejar, R.; Quintela-Fandino, M. Usefulness and real-world outcomes of next generation sequencing testing in patients with cancer: An observational study on the impact of selection based on clinical judgement. EClinicalMedicine 2023, 60, 102029. [Google Scholar] [CrossRef]
  281. Gulilat, M.; Lamb, T.; Teft, W.A.; Wang, J.; Dron, J.S.; Robinson, J.F.; Tirona, R.G.; Hegele, R.A.; Kim, R.B.; Schwarz, U.I. Targeted next generation sequencing as a tool for precision medicine. BMC Med. Genomics 2019, 12, 81. [Google Scholar] [CrossRef]
  282. Bai, J.W.; Qiu, S.Q.; Zhang, G.J. Molecular and functional imaging in cancer-targeted therapy: Current applications and future directions. Signal Transduct. Target. Ther. 2023, 8, 89. [Google Scholar] [CrossRef] [PubMed]
  283. Kircher, M.F.; Hricak, H.; Larson, S.M. Molecular imaging for personalized cancer care. Mol. Oncol. 2012, 6, 182–195. [Google Scholar] [CrossRef]
  284. Saini, K.S.; Twelves, C. Determining lines of therapy in patients with solid cancers: A proposed new systematic and comprehensive framework. Br. J. Cancer 2021, 125, 155–163. [Google Scholar] [CrossRef]
  285. Reddy, K.; Gharde, P.; Tayade, H.; Patil, M.; Reddy, L.S.; Surya, D. Advancements in Robotic Surgery: A Comprehensive Overview of Current Utilizations and Upcoming Frontiers. Cureus 2023, 15, e50415. [Google Scholar] [CrossRef]
  286. Mysorekar, V.V. Sentinel Lymph Node Biopsy in Breast Cancer. World J. Oncol. 2010, 1, e50415. [Google Scholar] [CrossRef]
  287. Anand, U.; Dey, A.; Chandel, A.K.S.; Sanyal, R.; Mishra, A.; Pandey, D.K.; De Falco, V.; Upadhyay, A.; Kandimalla, R.; Chaudhary, A.; et al. Cancer chemotherapy and beyond: Current status, drug candidates, associated risks and progress in targeted therapeutics. Genes. Dis. 2023, 10, 1367–1401. [Google Scholar] [CrossRef]
  288. Schirrmacher, V. From chemotherapy to biological therapy: A review of novel concepts to reduce the side effects of systemic cancer treatment (Review). Int. J. Oncol. 2019, 54, 407–419. [Google Scholar] [CrossRef]
  289. Li, X.; Yan, Z.; Kong, D.; Zou, W.; Wang, J.; Sun, D.; Jiang, Y.; Zheng, C. Erythropoiesis-stimulating agents in the management of cancer patients with anemia: A meta-analysis. Chin. J. Cancer Res. 2014, 26, 268–276. [Google Scholar] [CrossRef]
  290. Johnson-Wimbley, T.D.; Graham, D.Y. Diagnosis and management of iron deficiency anemia in the 21st century. Therap Adv. Gastroenterol. 2011, 4, 177–184. [Google Scholar] [CrossRef]
  291. Culy, C.R.; Spencer, C.M. Amifostine: An update on its clinical status as a cytoprotectant in patients with cancer receiving chemotherapy or radiotherapy and its potential therapeutic application in myelodysplastic syndrome. Drugs 2001, 61, 641–684. [Google Scholar] [CrossRef]
  292. Kciuk, M.; Yahya, E.B.; Mohamed Ibrahim Mohamed, M.; Rashid, S.; Iqbal, M.O.; Kontek, R.; Abdulsamad, M.A.; Allaq, A.A. Recent Advances in Molecular Mechanisms of Cancer Immunotherapy. Cancers 2023, 15, 2721. [Google Scholar] [CrossRef]
  293. Bayat Mokhtari, R.; Homayouni, T.S.; Baluch, N.; Morgatskaya, E.; Kumar, S.; Das, B.; Yeger, H. Combination therapy in combating cancer. Oncotarget 2017, 8, 38022–38043. [Google Scholar] [CrossRef]
  294. LaRiviere, M.J.; Santos, P.M.G.; Hill-Kayser, C.E.; Metz, J.M. Proton Therapy. Hematol. Oncol. Clin. N. Am. 2019, 33, 989–1009. [Google Scholar] [CrossRef]
  295. Waarts, M.R.; Stonestrom, A.J.; Park, Y.C.; Levine, R.L. Targeting mutations in cancer. J. Clin. Investig. 2022, 132, 1–11. [Google Scholar] [CrossRef]
  296. Maximiano, S.; Magalhães, P.; Guerreiro, M.P.; Morgado, M. Trastuzumab in the Treatment of Breast Cancer. BioDrugs 2016, 30, 75–86. [Google Scholar] [CrossRef]
  297. Welty, N.E.; Gill, S.I. Cancer Immunotherapy Beyond Checkpoint Blockade: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2022, 4, 563–578. [Google Scholar] [CrossRef]
  298. Guo, Z.; Zhang, R.; Yang, A.G.; Zheng, G. Diversity of immune checkpoints in cancer immunotherapy. Front. Immunol. 2023, 14, 1121285. [Google Scholar] [CrossRef]
  299. Liu, K.; Sun, Q.; Liu, Q.; Li, H.; Zhang, W.; Sun, C. Focus on immune checkpoint PD-1/PD-L1 pathway: New advances of polyphenol phytochemicals in tumor immunotherapy. Biomed. Pharmacother. 2022, 154, 113618. [Google Scholar] [CrossRef]
  300. Tang, T.; Huang, X.; Zhang, G.; Hong, Z.; Bai, X.; Liang, T. Advantages of targeting the tumor immune microenvironment over blocking immune checkpoint in cancer immunotherapy. Signal Transduct. Target. Ther. 2021, 6, 72. [Google Scholar] [CrossRef]
  301. Sharma, P.; Allison, J.P. The future of immune checkpoint therapy. Science 2015, 348, 56–61. [Google Scholar] [CrossRef]
  302. Karasarides, M.; Cogdill, A.P.; Robbins, P.B.; Bowden, M.; Burton, E.M.; Butterfield, L.H.; Cesano, A.; Hammer, C.; Haymaker, C.L.; Horak, C.E.; et al. Hallmarks of Resistance to Immune-Checkpoint Inhibitors. Cancer Immunol. Res. 2022, 10, 372–383. [Google Scholar] [CrossRef]
  303. Deckers, J.; Anbergen, T.; Hokke, A.M.; de Dreu, A.; Schrijver, D.P.; de Bruin, K.; Toner, Y.C.; Beldman, T.J.; Spangler, J.B.; de Greef, T.F.A.; et al. Engineering cytokine therapeutics. Nat. Rev. Bioeng. 2023, 1, 286–303. [Google Scholar] [CrossRef]
  304. Yi, M.; Li, T.; Niu, M.; Zhang, H.; Wu, Y.; Wu, K.; Dai, Z. Targeting cytokine and chemokine signaling pathways for cancer therapy. Signal Transduct. Target. Ther. 2024, 9, 176. [Google Scholar] [CrossRef]
  305. Bulgarelli, J.; Piccinini, C.; Petracci, E.; Pancisi, E.; Granato, A.M.; de Rosa, F.; Guidoboni, M.; Petrini, M.; Ancarani, V.; Foschi, G.; et al. Radiotherapy and High-Dose Interleukin-2: Clinical and Immunological Results of a Proof of Principle Study in Metastatic Melanoma and Renal Cell Carcinoma. Front. Immunol. 2021, 12, 778459. [Google Scholar] [CrossRef]
  306. Saxton, R.A.; Glassman, C.R.; Garcia, K.C. Emerging principles of cytokine pharmacology and therapeutics. Nat. Rev. Drug Discov. 2023, 22, 21–37. [Google Scholar] [CrossRef]
  307. Cai, M.; Huang, X.; Huang, X.; Ju, D.; Zhu, Y.Z.; Ye, L. Research progress of interleukin-15 in cancer immunotherapy. Front. Pharmacol. 2023, 14, 1184703. [Google Scholar] [CrossRef]
  308. Uscanga-Palomeque, A.C.; Chavez-Escamilla, A.K.; Alvizo-Baez, C.A.; Saavedra-Alonso, S.; Terrazas-Armendariz, L.D.; Tamez-Guerra, R.S.; Rodriguez-Padilla, C.; Alcocer-Gonzalez, J.M. CAR-T Cell Therapy: From the Shop to Cancer Therapy. Int. J. Mol. Sci. 2023, 24, 15688. [Google Scholar] [CrossRef]
  309. Sterner, R.C.; Sterner, R.M. CAR-T cell therapy: Current limitations and potential strategies. Blood Cancer J. 2021, 11, 69. [Google Scholar] [CrossRef]
  310. Luo, J.; Zhang, X. Challenges and innovations in CAR-T cell therapy: A comprehensive analysis. Front. Oncol. 2024, 14, 1399544. [Google Scholar] [CrossRef]
  311. Fan, T.; Zhang, M.; Yang, J.; Zhu, Z.; Cao, W.; Dong, C. Therapeutic cancer vaccines: Advancements, challenges, and prospects. Signal Transduct. Target. Ther. 2023, 8, 450. [Google Scholar] [CrossRef]
  312. Xie, N.; Shen, G.; Gao, W.; Huang, Z.; Huang, C.; Fu, L. Neoantigens: Promising targets for cancer therapy. Signal Transduct. Target. Ther. 2023, 8, 9. [Google Scholar] [CrossRef]
  313. Zanotta, S.; Galati, D.; De Filippi, R.; Pinto, A. Enhancing Dendritic Cell Cancer Vaccination: The Synergy of Immune Checkpoint Inhibitors in Combined Therapies. Int. J. Mol. Sci. 2024, 25, 7509. [Google Scholar] [CrossRef] [PubMed]
  314. Lin, D.; Shen, Y.; Liang, T. Oncolytic virotherapy: Basic principles, recent advances and future directions. Signal Transduct. Target. Ther. 2023, 8, 156. [Google Scholar] [CrossRef]
  315. Bommareddy, P.K.; Patel, A.; Hossain, S.; Kaufman, H.L. Talimogene Laherparepvec (T-VEC) and Other Oncolytic Viruses for the Treatment of Melanoma. Am. J. Clin. Dermatol. 2017, 18, 1–15. [Google Scholar] [CrossRef]
  316. Lemos de Matos, A.; Franco, L.S.; McFadden, G. Oncolytic Viruses and the Immune System: The Dynamic Duo. Mol. Ther. Methods Clin. Dev. 2020, 17, 349–358. [Google Scholar] [CrossRef]
  317. Kaufman, H.L.; Shalhout, S.Z.; Iodice, G. Talimogene Laherparepvec: Moving From First-In-Class to Best-In-Class. Front. Mol. Biosci. 2022, 9, 834841. [Google Scholar] [CrossRef]
  318. Tsai, C.C.; Yang, Y.S.H.; Chen, Y.F.; Huang, L.Y.; Yang, Y.N.; Lee, S.Y.; Wang, W.L.; Lee, H.L.; Whang-Peng, J.; Lin, H.Y.; et al. Integrins and Actions of Androgen in Breast Cancer. Cells 2023, 12, 2126. [Google Scholar] [CrossRef]
  319. Cesur-Ergun, B.; Demir-Dora, D. Gene therapy in cancer. J. Gene Med. 2023, 25, e3550. [Google Scholar] [CrossRef]
  320. Konda, P.; Garinet, S.; Van Allen, E.M.; Viswanathan, S.R. Genome-guided discovery of cancer therapeutic targets. Cell Rep. 2023, 42, 112978. [Google Scholar] [CrossRef]
  321. Liu, B.; Zhou, H.; Tan, L.; Siu, K.T.H.; Guan, X.Y. Exploring treatment options in cancer: Tumor treatment strategies. Signal Transduct. Target. Ther. 2024, 9, 175. [Google Scholar] [CrossRef]
  322. Nyquist, M.D.; Corella, A.; Coleman, I.; De Sarkar, N.; Kaipainen, A.; Ha, G.; Gulati, R.; Ang, L.; Chatterjee, P.; Lucas, J.; et al. Combined TP53 and RB1 Loss Promotes Prostate Cancer Resistance to a Spectrum of Therapeutics and Confers Vulnerability to Replication Stress. Cell Rep. 2020, 31, 107669. [Google Scholar] [CrossRef]
  323. Wang, J.H.; Gessler, D.J.; Zhan, W.; Gallagher, T.L.; Gao, G. Adeno-associated virus as a delivery vector for gene therapy of human diseases. Signal Transduct. Target. Ther. 2024, 9, 78. [Google Scholar] [CrossRef]
  324. Sheikh, S.; Ernst, D.; Keating, A. Prodrugs and prodrug-activated systems in gene therapy. Mol. Ther. 2021, 29, 1716–1728. [Google Scholar] [CrossRef]
  325. Fang, H.; Yan, H.H.N.; Bilardi, R.A.; Flensburg, C.; Yang, H.; Barbour, J.A.; Siu, H.C.; Turski, M.; Chew, E.; Xu, Z.; et al. Ganciclovir-induced mutations are present in a diverse spectrum of post-transplant malignancies. Genome Med. 2022, 14, 124. [Google Scholar] [CrossRef]
  326. Rabaan, A.A.; AlSaihati, H.; Bukhamsin, R.; Bakhrebah, M.A.; Nassar, M.S.; Alsaleh, A.A.; Alhashem, Y.N.; Bukhamseen, A.Y.; Al-Ruhimy, K.; Alotaibi, M.; et al. Application of CRISPR/Cas9 Technology in Cancer Treatment: A Future Direction. Curr. Oncol. 2023, 30, 1954–1976. [Google Scholar] [CrossRef]
  327. Sayed, N.; Allawadhi, P.; Khurana, A.; Singh, V.; Navik, U.; Pasumarthi, S.K.; Khurana, I.; Banothu, A.K.; Weiskirchen, R.; Bharani, K.K. Gene therapy: Comprehensive overview and therapeutic applications. Life Sci. 2022, 294, 120375. [Google Scholar] [CrossRef]
  328. Zugazagoitia, J.; Guedes, C.; Ponce, S.; Ferrer, I.; Molina-Pinelo, S.; Paz-Ares, L. Current Challenges in Cancer Treatment. Clin. Ther. 2016, 38, 1551–1566. [Google Scholar] [CrossRef]
  329. Wu, X.; Yang, X.; Dai, Y.; Zhao, Z.; Zhu, J.; Guo, H.; Yang, R. Single-cell sequencing to multi-omics: Technologies and applications. Biomark. Res. 2024, 12, 110. [Google Scholar] [CrossRef]
  330. Bou Antoun, N.; Chioni, A.M. Dysregulated Signalling Pathways Driving Anticancer Drug Resistance. Int. J. Mol. Sci. 2023, 24, 12222. [Google Scholar] [CrossRef]
  331. West, J.; You, L.; Zhang, J.; Gatenby, R.A.; Brown, J.S.; Newton, P.K.; Anderson, A.R.A. Towards Multidrug Adaptive Therapy. Cancer Res. 2020, 80, 1578–1589. [Google Scholar] [CrossRef]
  332. Min, H.Y.; Lee, H.Y. Molecular targeted therapy for anticancer treatment. Exp. Mol. Med. 2022, 54, 1670–1694. [Google Scholar] [CrossRef]
  333. Sabnis, A.J.; Bivona, T.G. Principles of Resistance to Targeted Cancer Therapy: Lessons from Basic and Translational Cancer Biology. Trends Mol. Med. 2019, 25, 185–197. [Google Scholar] [CrossRef]
  334. Attili, I.; Corvaja, C.; Spitaleri, G.; Del Signore, E.; Trillo Aliaga, P.; Passaro, A.; de Marinis, F. New Generations of Tyrosine Kinase Inhibitors in Treating NSCLC with Oncogene Addiction: Strengths and Limitations. Cancers 2023, 15, 5079. [Google Scholar] [CrossRef]
  335. Labrie, M.; Brugge, J.S.; Mills, G.B.; Zervantonakis, I.K. Therapy resistance: Opportunities created by adaptive responses to targeted therapies in cancer. Nat. Rev. Cancer 2022, 22, 323–339. [Google Scholar] [CrossRef]
  336. Camerini, A.; Mazzoni, F.; Scotti, V.; Tibaldi, C.; Sbrana, A.; Calabro, L.; Caliman, E.; Ciccone, L.P.; Bernardini, L.; Graziani, J.; et al. Efficacy and Safety of Chemotherapy after Immunotherapy in Patients with Advanced Non-Small-Cell Lung Cancer. J. Clin. Med. 2024, 13, 3642. [Google Scholar] [CrossRef]
  337. Belluomini, L.; Avancini, A.; Sposito, M.; Pontolillo, L.; Tregnago, D.; Trestini, I.; Insolda, J.; Carbognin, L.; Milella, M.; Bria, E.; et al. Integrating nutrition, physical exercise, psychosocial support and antiemetic drugs into CINV management: The road to success. Crit. Rev. Oncol. Hematol. 2024, 201, 104444. [Google Scholar] [CrossRef]
  338. Chen, D.; Liu, X.; Lu, X.; Tian, J. Nanoparticle drug delivery systems for synergistic delivery of tumor therapy. Front. Pharmacol. 2023, 14, 1111991. [Google Scholar] [CrossRef]
Figure 1. A variety of immunotherapies are employed to treat cancer, including immune-checkpoint inhibitors, cancer vaccines, cytokines, viruses, and adoptive cell transfer.
Figure 1. A variety of immunotherapies are employed to treat cancer, including immune-checkpoint inhibitors, cancer vaccines, cytokines, viruses, and adoptive cell transfer.
Biomedicines 12 02659 g001
Figure 2. Schematic depiction of diagnostics and therapy for malignant tumors. The cancer development pathway includes three stages. First, genetic and epigenetic mutations in oncogenes (e.g., RAS and MYC) and tumor suppressor genes (e.g., TP53 and BRCA1) are shown in the initiation step. The second step shows the illustrated mechanisms like evasion of apoptosis, sustained angiogenesis, and immune evasion within the tumor microenvironment. Finally, visualize invasion and metastasis processes, with tumor cells spreading through the blood or lymphatic systems in the metastasis step. The diagnostic tools are highlight imaging techniques (e.g., CT, MRI, and PET) and molecular diagnostics (e.g., biomarkers and liquid biopsies) as they aid early detection and monitoring. Therapeutic interventions involve various therapeutic strategies. One is traditional therapies that include surgery, chemotherapy, and radiation therapy. Another is targeted therapies that include tyrosine kinase inhibitors and monoclonal antibodies. The other is immunotherapy that includes immune checkpoint inhibitors and CAR T-cell therapy.
Figure 2. Schematic depiction of diagnostics and therapy for malignant tumors. The cancer development pathway includes three stages. First, genetic and epigenetic mutations in oncogenes (e.g., RAS and MYC) and tumor suppressor genes (e.g., TP53 and BRCA1) are shown in the initiation step. The second step shows the illustrated mechanisms like evasion of apoptosis, sustained angiogenesis, and immune evasion within the tumor microenvironment. Finally, visualize invasion and metastasis processes, with tumor cells spreading through the blood or lymphatic systems in the metastasis step. The diagnostic tools are highlight imaging techniques (e.g., CT, MRI, and PET) and molecular diagnostics (e.g., biomarkers and liquid biopsies) as they aid early detection and monitoring. Therapeutic interventions involve various therapeutic strategies. One is traditional therapies that include surgery, chemotherapy, and radiation therapy. Another is targeted therapies that include tyrosine kinase inhibitors and monoclonal antibodies. The other is immunotherapy that includes immune checkpoint inhibitors and CAR T-cell therapy.
Biomedicines 12 02659 g002
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

Tsai, C.-C.; Wang, C.-Y.; Chang, H.-H.; Chang, P.T.S.; Chang, C.-H.; Chu, T.Y.; Hsu, P.-C.; Kuo, C.-Y. Diagnostics and Therapy for Malignant Tumors. Biomedicines 2024, 12, 2659. https://doi.org/10.3390/biomedicines12122659

AMA Style

Tsai C-C, Wang C-Y, Chang H-H, Chang PTS, Chang C-H, Chu TY, Hsu P-C, Kuo C-Y. Diagnostics and Therapy for Malignant Tumors. Biomedicines. 2024; 12(12):2659. https://doi.org/10.3390/biomedicines12122659

Chicago/Turabian Style

Tsai, Chung-Che, Chun-Yu Wang, Hsu-Hung Chang, Phebe Ting Syuan Chang, Chuan-Hsin Chang, Tin Yi Chu, Po-Chih Hsu, and Chan-Yen Kuo. 2024. "Diagnostics and Therapy for Malignant Tumors" Biomedicines 12, no. 12: 2659. https://doi.org/10.3390/biomedicines12122659

APA Style

Tsai, C. -C., Wang, C. -Y., Chang, H. -H., Chang, P. T. S., Chang, C. -H., Chu, T. Y., Hsu, P. -C., & Kuo, C. -Y. (2024). Diagnostics and Therapy for Malignant Tumors. Biomedicines, 12(12), 2659. https://doi.org/10.3390/biomedicines12122659

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