Next Article in Journal
Ethylenediaminetetraacetic Acid (EDTA)-Decalcified, Formalin-Fixed Paraffin-Embedded (FFPE) Tumor Tissue Shows Comparable Quality and Quantity of DNA to Non-Decalcified Tissue in Next-Generation Sequencing (NGS)
Previous Article in Journal
ERBB2 Mutation Testing in NSCLC: A Pan-European Real-World Evaluation of the Oncomine Precision Assay
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Future of Cancer Diagnosis and Treatment: Unlocking the Power of Biomarkers and Personalized Molecular-Targeted Therapies

1
Institute of Biotechnology, Addis Ababa University, Addis Ababa 1000, Ethiopia
2
College of Veterinary Medicine, Jigjiga University, Jigjiga 1020, Ethiopia
3
Health Biotechnology Directorate, Bio and Emerging Technology Institute, Addis Ababa 5954, Ethiopia
*
Authors to whom correspondence should be addressed.
J. Mol. Pathol. 2025, 6(3), 20; https://doi.org/10.3390/jmp6030020
Submission received: 28 March 2025 / Revised: 7 May 2025 / Accepted: 15 May 2025 / Published: 28 August 2025

Abstract

Cancer remains a leading global health challenge, with conventional diagnostic and treatment methods often lacking precision and adaptability. This review explores transformative advancements that are reshaping oncology by addressing these limitations. It begins with an overview of cancer’s complexity, emphasizing the shortcomings of conventional tools such as imaging and chemotherapy, which frequently fail to deliver targeted care. The discussion then shifts to biomarkers, which represent a groundbreaking frontier in early detection, enabling the identification of unique biological signatures that signal the presence of cancer with heightened sensitivity. Building on this foundation, the review examines personalized molecular therapies, which target the specific genetic and molecular vulnerabilities of tumors. These therapies not only enhance treatment efficacy but also minimize adverse effects, offering patients improved outcomes and quality of life. By integrating biomarker-driven diagnostics with tailored therapeutic strategies, a new paradigm of precision oncology emerges, bridging the gap between early detection and effective intervention. Real-world case studies highlight both successes, such as significantly improved survival rates, and persistent challenges, including accessibility and cost barriers. Looking ahead, the review outlines pathways by which to scale these innovations, emphasizing the critical need for robust infrastructure, sustained research investment, and equitable healthcare policies. It concludes by envisioning a future where biomarkers and personalized therapies converge to redefine cancer care, offering earlier detection, precise interventions, and better patient experiences. This work underscores the urgency of adopting cutting-edge approaches to overcome cancer’s persistent threats, paving the way for a more effective and humane era in oncology.

1. Introduction

Cancer is a relentless enemy, a disease where cells go rogue, growing and dividing without the usual checks and balances that keep our bodies in harmony. Normally, cells follow a tight script: they grow when needed, divide to replace old ones, and die off through a process called apoptosis when their job is complete [1]. However, in cancer, this script becomes scrambled, sometimes by genetic mutations passed down through families, other times by causes such as cigarette smoke, UV rays, or viruses like human papillomavirus (HPV) [2]. The result is that cells multiply uncontrollably, forming tumors that can stay local or, in the worst cases, break away and spread throughout the body in a process called metastasis [3]. This is a global crisis, claiming around 10 million lives each year, according to the World Health Organization (WHO). Additionally, the burden is not shared equally, 70% of these deaths happen in low- and middle-income countries, where spotting cancer early or treating it effectively often feels like a distant dream [4].
For too long, cancer diagnosis has relied on tools that often fall short, symptoms, like fatigue or a persistent cough, are easily dismissed, and, even when concerns arise, traditional tests can be unreliable, either producing false alarms or failing to detect real cancerous threats. However, the future of cancer care is on the brink of a revolution, one that is driven by biomarkers and personalized molecular-targeted therapies. These advancements promise to catch cancer sooner and fight it smarter [5,6].
Traditionally, cancer detection has involved a patient noticing vague symptoms, leading to a series of tests, such as X-rays and biopsies. Unfortunately, the disease often remains silent in its early stages, making it challenging to treat once detected [7]. This is where biomarkers come in, transforming how we identify cancer. A simple blood draw can reveal the disease before symptoms appear, using plasma proteomics to spot patterns unique to cancer. These protein signatures indicate not only the presence of cancer but also its type and behavior. Researchers are already using this approach to pinpoint lung cancer in smokers or breast cancer in women with unclear mammograms. Moreover, biomarkers can flag which patients will respond to certain drugs, ensuring that the right therapy is given faster [8,9].
However, finding cancer early is only half the battle; beating it is where personalized molecular-targeted therapies shine. In the past, we have thrown everything at cancer—surgery to cut it out, radiation to zap it, chemotherapy to poison it—hoping something sticks. All of these shotgun approaches can work, but they often hit healthy cells too, leaving patients exhausted, sick, and sometimes no better off [10,11]. As an example, all breast cancer patients were in the past treated with a combination of surgery, chemotherapy, and radiation, regardless of their tumor types; however, researchers discovered that some breast cancers overexpress the human epidermal growth factor receptor 2 (HER2) protein, which promotes rapid tumor growth. Instead of using traditional chemotherapy alone, targeted drugs like trastuzumab (Herceptin) and pertuzumab (Perjeta) have been developed to specifically block HER2 signaling [12]. This breakthrough dramatically improved survival rates for HER2-positive breast cancer patients, while reducing the side effects associated with traditional chemotherapy. This exemplifies how personalized therapies can attack cancer more precisely, leading to better outcomes [13].
Molecular-targeted therapies function like a precision tool, focusing on the specific genetic traits driving a tumor. They are built on the idea that every cancer has a molecular weak spot, a mutated gene or overactive protein that keeps it alive [14]. Drugs like imatinib, for example, target a specific protein in chronic myeloid leukemia, turning a once-deadly disease into something people live with for decades [15]. Additionally, a simple blood test might reveal that a lung cancer patient has an epidermal growth factor receptor (EGFR) mutation, allowing doctors to bypass chemotherapy and prescribe a targeted drug like osimertinib, which directly targets the mutation. Real-world evidence supports this approach; a recent cervical cancer trial found that adding a brief round of chemotherapy before standard treatment reduced mortality by 40%, highlighting the potential of tweaking therapy based on a tumor’s unique profile [16,17].
This integrated approach is not science fiction; it is already happening and advancing rapidly [18]. Scientists are racing to map more biomarkers, from deoxyribonucleic acid (DNA) fragments to tiny vesicles released by cells, which can detect cancer early or predict treatment responses. Consider melanoma, once a death sentence, now often treatable with drugs like pembrolizumab, which boosts the immune response against cancer cells flagged by biomarkers [19]. Additionally, artificial intelligence (AI) is analyzing data from millions of patients to identify biomarker patterns that might be missed by humans. The goal is to ensure that advancements in cancer care reach everyone, from farmers in rural Ethiopia to factory workers in Ohio [20,21].
This review offers a front-row seat for the future of cancer care. We explored how biomarkers and personalized molecular-targeted therapies are transforming treatment, from the detection of cancer in its early stages to the design of therapies that match a tumor’s unique genetic makeup. These are real, life-saving tools, and there are even more on the horizon. We highlighted therapies that directly target cancer, supported by stories of patients who have gone from hopeless to thriving. Finally, we looked ahead to how these innovations could reduce cancer’s global impact and shift the focus from treatment to prevention. By unlocking the potential of biomarkers and targeted therapies, we are not just fighting back; we are creating a future where cancer no longer wins.

2. The Limits of Today’s Cancer Diagnosis and Treatment Toolbox

Cancer remains one of the most complex and challenging diseases to diagnose and treat. Despite significant advancements in medical science, technology, and treatment strategies, there are still substantial limitations in today’s cancer diagnosis and treatment toolbox. These limitations span several areas, including early detection, personalized medicine, treatment efficacy, side effects, and accessibility [7,22]. One major challenge is cancer of unknown primary (CUP), which accounts for 2.3% to 7.8% of all malignant tumors and ranks as a leading cause of cancer-related deaths. Effective detection methods for CUP are lacking, with 20–50% of patients never having their primary site identified [23,24]. This highlights the urgent need for more advanced diagnostic approaches to accurately determine the tumor’s origin and tissue type [25].
Many conventional diagnostic tools lack accuracy, often failing to identify cancer in its earliest stages or provide a complete understanding of tumor biology (Table 1) [26]. Patients may experience vague symptoms like fatigue, a persistent cough, or mild discomfort, which can easily be mistaken for stress or common ailments. By the time symptoms become severe enough to raise concern, the disease may have already spread, turning a once-treatable condition into a life-threatening battle. This reliance on late-stage symptoms and insufficiently sensitive tests means we often detect cancer too late, when treatment options are more limited and less effective [27,28].
When cancer is suspected, diagnosing it with precision remains a challenge. Doctors rely on various tools, such as blood tests, X-rays, CT scans, and biopsies, but each has limitations [29]. For example, mammograms, the standard screening method for breast cancer, fail to detect about one in five cases in women with dense breast tissue. Blood tests like the prostate-specific antigen (PSA) test for prostate cancer can indicate potential issues but are notorious for false positives, leading to unnecessary anxiety and invasive follow-ups [30]. A 2020 UK study revealed that 30% of ovarian cancer cases were initially misdiagnosed, as vague symptoms were mistaken for less serious conditions [31]. Limited access to advanced imaging further contributes to late-stage detection, highlighting the need for more reliable early-stage diagnostic methods [32].
False negatives can also be dangerous. A biopsy that samples the wrong area may falsely reassure a patient while cancer continues to spread undetected. Some tumors, particularly in the pancreas or ovaries, are especially elusive, hiding deep within the body, where routine screenings struggle to reach [33]. A well-known example is the case of actor Patrick Swayze, who was diagnosed with pancreatic cancer only after it had already advanced to stage IV, despite experiencing symptoms months earlier [34]. Similarly, many women with ovarian cancer receive late diagnoses because early symptoms are mistaken for common digestive issues [35]. Furthermore, access to specialists and advanced imaging is limited in rural clinics, leading to delays in diagnosis or missed opportunities for early detection [36].
Cancer treatment presents another major challenge, as current diagnostic and therapeutic tools often fall short. While treatment strategies have become more advanced, they still face hurdles, such as drug resistance, severe side effects, and limited effectiveness across different cancer types. The standard approach has traditionally relied on surgery, radiation, and chemotherapy [37,38]. Surgery can be highly effective if cancer is caught early and remains localized. for instance, removing a small breast tumor before it spreads to the lymph nodes. However, once cancer metastasizes, surgery alone is rarely enough. On the other hand, radiation therapy uses high-energy beams to shrink tumors or slow their uncontrolled growth, but it also damages healthy tissue, leaving patients fatigued or permanently scarred [39,40]. The other treatment option, chemotherapy, the most aggressive approach, targets rapidly dividing cells, both cancerous and healthy. While it has saved countless lives, its side effects can be brutal and can include hair loss, nausea, and immune suppression, with only a fraction of patients experiencing significant benefit [41,42]. The numbers starkly reveal the limitations of current cancer diagnosis and treatment.
These conventional treatments follow “a one-size-fits-all” approach, attacking cancer broadly without considering its unique genetic and molecular characteristics. Currently, emerging therapies like immunotherapy and precision medicine offer more targeted options by harnessing the immune system or tailoring treatments to specific genetic mutations. However, these innovations remain expensive, often inaccessible in many regions, and inconsistent in their success rates, leaving many patients without viable options [43,44]. For example, a farmer in rural Ethiopia may experience persistent stomach pain for months before being diagnosed with a gastric cancer that has already spread, due in part to a lack of routine screenings. Even in wealthier countries, survival rates for certain cancers remain dismally low; pancreatic cancer, for instance, is rarely detected before reaching stage IV, leading to a five-year survival rate of just 10% [45].
These disparities are not just medical; they are deeply personal. Families worldwide watch loved ones succumb to cancer not because the disease is invincible, but because our current detection and treatment methods often fail. The reality is frustrating: we are battling a highly adaptable enemy with tools that can be outdated and imprecise. The urgent need for sharper, more personalized approaches is clear, and the next wave of innovation may finally provide the breakthroughs we have been waiting for [46].
Table 1. Cancer diagnosis: Conventional vs. advanced methods [47].
Table 1. Cancer diagnosis: Conventional vs. advanced methods [47].
AspectConventional DiagnosisAdvanced Diagnosis
Techniques
  • Biopsy (tissue examination)
  • Imaging (X-ray, CT, MRI, ultrasound)
  • Blood tests (tumor markers like PSA, CA-125)
  • Liquid biopsy (circulating tumor DNA, CTCs)
  • Next-generation sequencing (NGS)
  • AI-powered imaging analysis
  • Single-cell RNA sequencing (scRNA-seq)
  • Multi-omics profiling (genomics, proteomics, metabolomics)
AccuracyModerate, requires invasive procedures and may not detect early-stage cancerHigh, enables early detection and real-time monitoring of tumor evolution
Personalization“One-size-fits-all” approach, based on histology and general tumor characteristicsHighly personalized, identifying genetic mutations and molecular signatures for tailored treatment
Speed of ResultsCan take weeks due to tissue processing and pathologyFaster, especially with AI-driven and liquid biopsy techniques

3. Biomarkers: The New Frontier in Cancer Detection

Cancer remains a major global health challenge, highlighting the urgent need for continuous advancements in diagnostic and treatment strategies. Despite significant progress in cancer care, many treatment options are still limited by the inability to detect cancer in its early stages, when it is most treatable. Therefore, improving early detection methods, refining treatment precision, and expanding access to innovative therapies are critical steps toward reducing cancer-related mortality and improving patient outcomes worldwide [46,48].
One of the most promising developments in cancer research is the rapid evolution of biomarker technology, which is transforming how we detect and diagnose cancer. Biomarkers are biological molecules found in blood, urine, or tissue samples that provide a way to identify the presence of cancer long before symptoms appear, enabling earlier intervention that can significantly improve patient outcomes. Unlike conventional detection approaches and technologies, such as imaging and biopsies, which often identify cancer at later stages when treatment options are more limited, biomarkers can detect malignancies at their initiation [19,29,49]. This approach not only allows for more timely treatment but also offers a non-invasive or minimally invasive means of monitoring the disease [50,51].
Biomarkers include things such as enzyme levels, protein expressions, fragments of DNA, and other cellular signals that cancer leaves behind as it grows (Figure 1). Think of biomarkers as a tumor’s unique fingerprints, circulating in the blood, urine, or tissue, waiting to reveal their secrets if we know where and how to look [18,52].
Biomarkers not only indicate that something is wrong but also provide detailed insight, helping to identify the type of cancer, its location, and even its potential aggressiveness. Microsatellite instability-high (MSI-H) is a crucial biomarker in specific colorectal cancers. For instance, tumors exhibiting MSI-H status tend to respond well to immunotherapy, particularly pembrolizumab, resulting in significantly improved survival rates for patients, and in turn highlighting the importance of biomarker-driven treatment strategies [53,54].
Cancer biomarkers can be classified into four main types (Table 2), each playing a crucial role in the detection, management, and treatment of the disease [51]. Among these, diagnostic biomarkers are essential for early detection, often signaling the presence of cancer before symptoms appear. For example, prostate-specific antigen (PSA) for prostate cancer and cancer antigen 125 (CA-125) for ovarian cancer enable earlier intervention, improving treatment options [28,55].
Another significant category of biomarkers is that of prognostic biomarkers, which provide valuable insights into the aggressiveness of a cancer and can help predict its likely progression. By offering these insights, prognostic biomarkers allow doctors to assess the severity of the disease and make informed decisions regarding treatment plans. For instance, the presence of specific biomarkers can indicate whether the cancer is likely to spread quickly, enabling doctors to tailor more aggressive treatments when necessary [56,57].
The third type of biomarker is that of predictive biomarkers, which are crucial for determining how well a patient might respond to specific therapies. An example of this is HER2 in breast cancer, which helps guide the use of targeted treatments like trastuzumab (Herceptin). By specifically targeting cells that overexpress the HER2 protein, these predictive biomarkers increase the likelihood of treatment success [57,58].
Finally, monitoring biomarkers are essential for tracking the effectiveness of treatment over time and detecting any potential recurrence of cancer. By utilizing these biomarkers, they allow healthcare providers to adjust therapies as needed, ensuring patients receive optimal care [59,60].
Table 2. Types of biomarkers and their purpose [61].
Table 2. Types of biomarkers and their purpose [61].
Biomarker TypesPurpose Examples
Diagnostic biomarker Detect cancer presence PSA, CA-125, circulating tumor cells (CTCs)
Prognostic biomarker Predict cancer outcome HER2, Ki-67, tumor protein p53 (TP53) mutations
Predictive biomarker Guide treatment selection EGFR mutations, Programmed death-ligand 1 (PD-L1) expression
Monitoring biomarker Track disease progression Carcinoembryonic antigen (CEA), ctDNA, imaging-based markers
The study of cancer biomarkers is full of exciting possibilities, particularly in plasma proteomics, which analyzes thousands of proteins in blood to identify those signaling the presence of cancer (Table 3). Additionally, circulating tumor DNA (ctDNA) reveals genetic material released by tumors into the bloodstream, offering insights into tumor behavior [8,62]. Other biomarkers, such as ribonucleic acid (RNA) and exosomes (tiny vesicles released by cells), enhance our understanding of cancer. The tools used to analyze biomarkers are becoming increasingly sophisticated. Machine learning algorithms can identify patterns and trends that might otherwise go unnoticed, providing doctors with valuable information to guide treatment decisions [63,64].
The benefits of biomarkers are significant: they enable faster, less invasive, and more precise cancer detection compared with traditional biopsies and scans. By pinpointing specific mutations and characteristics, biomarkers not only help identify cancer but also provide a detailed roadmap for treatment, marking a new era in cancer care [52,61].
Evidence of the effectiveness of biomarkers is mounting. For instance, tests like carbohydrate-deficient transferrin (early CDT-lung) utilize a panel of protein biomarkers to detect lung cancer early, identifying suspicious cases up to a year before traditional CT scans [65,66]. Similarly, the blood test Septin9 for colorectal cancer identifies ctDNA with over 80% accuracy, serving as a less invasive alternative to colonoscopy. In prostate cancer, newer biomarker panels like the 4Kscore improve accuracy by combining multiple proteins, minimizing unnecessary procedures and ultimately reducing patient anxiety [67,68,69].
Table 3. List of various cancer types along with their associated biomarker types [70].
Table 3. List of various cancer types along with their associated biomarker types [70].
Cancer TypesBiomarker Types Monitoring Biomarker
Prostate cancerDiagnostic (PSA)Monitoring PSA levels
Ovarian cancerDiagnostic: Cancer antigen 125 (CA-125)Monitoring: CA-125 levels
Breast cancerPredictive: Human epidermal growth factor receptor 2 (HER2)Monitoring: HER2 levels
Lung cancerDiagnostic: Carbohydrate-deficient transferrin (early CDT-lung)Monitoring: CTDNA
Colorectal cancerDiagnostic: Carcinoembryonic antigen (CEA)Monitoring: CEA levels
MelanomaPrognostic: (BRAF mutations)Monitoring: Lactate dehydrogenase (LDH) levels
Pancreatic cancerDiagnostic: Carbohydrate antigen 19-9 (CA 19-9)Monitoring: CA 19-9 levels
Liver cancerDiagnostic: Alpha-fetoprotein (AFP)Monitoring: AFP levels
Bladder cancerDiagnostic: Nuclear matrix protein 22 (NMP22)Monitoring: NMP22 levels
Testicular cancerDiagnostic: Alpha-fetoprotein (AFP), human chorionic gonadotropin (hCG)Monitoring: AFP levels
In breast cancer, the Oncotype DX test analyzes tumor RNA to evaluate recurrence risk, enabling more informed treatment decisions. These advancements reflect a broader trend in personalized medicine, where biomarker testing plays a crucial role in tailoring treatments to individual needs [71]. Furthermore, they offer a glimpse into a future where a simple blood draw could detect cancers like pancreatic cancer before they become fatal or identify ovarian cancer before misdiagnosis occurs [72,73].
This new frontier is exciting not only for its earlier detection but also for its greater accuracy. While biomarkers are not yet flawless—some tests can be costly and others require further refinement—the progress is undeniable. Researchers are diligently working to discover more markers, combining findings with advanced technologies like artificial intelligence to uncover patterns that might otherwise go unnoticed [74]. The ultimate vision is a future where cancer screening is as routine as an annual check-up, catching the disease early enough that it does not stand a chance. With ongoing advancements, we are moving closer to a world where early cancer detection becomes the norm rather than the exception [75].

4. Personalized Molecular Therapies: Targeting Cancer’s Weak Spots

Cancer is a complex and heterogeneous disease, marked by uncontrolled cell growth that is in turn driven by genetic mutations, epigenetic alterations, and dysregulated signaling pathways. Traditional treatments, like chemotherapy and radiation, often target rapidly dividing cells indiscriminately, resulting in significant side effects and limited effectiveness for some patients. However, recent advancements in genomics, transcriptomics, proteomics, metabolomics, and computational biology have opened the door to personalized molecular therapies. These therapies aim to target specific vulnerabilities within individual tumors by identifying and exploiting their unique genetic and molecular characteristics, ultimately enhancing treatment efficacy [11,76].
For a long time, cancer treatment has been a blunt instrument, but we are entering a new era with personalized molecular-targeted therapies that directly address the disease’s underlying mechanisms. At the core of personalized molecular therapy is genomic profiling, which involves analyzing the DNA, RNA, and proteins of a patient’s tumor to identify actionable mutations or biomarkers. Advanced techniques such as next-generation sequencing (NGS), whole-exome sequencing (WES), and transcriptomics facilitate a comprehensive molecular characterization of tumors [10,77].
Next-generation sequencing (NGS) is a rapidly evolving technology that analyzes DNA or RNA extracted from tumor samples, enabling the identification of actionable mutations. Notably, alterations in genes such as breast cancer genes 1 (BRCA1) and 2 (BRCA2) are crucial, as they can significantly influence treatment decisions and the use of targeted therapies. For example, in breast cancer, specific BRCA mutations can lead to the application of poly (ADP-ribose) polymerase (PARP) inhibitors, which effectively target tumors with these genetic changes. Thus, integrating NGS into clinical practice enhances the precision of cancer treatment and paves the way for personalized medicine [78,79].
Building on this foundation, gene expression profiling, utilizing techniques like RNA sequencing and microarrays, plays a vital role in measuring gene activity to classify tumors into distinct molecular subtypes. By analyzing gene activity patterns, clinicians can make informed decisions about treatment options, tailoring therapies to the unique molecular features of each tumor. Consequently, this approach not only improves the precision of cancer care but also optimizes treatment strategies, ultimately enhancing patient outcomes [80,81].
In addition to these advancements, proteomics, which employs mass spectrometry, is essential for quantifying protein biomarkers like HER2 in breast cancer. By measuring the levels of these proteins, clinicians can gain insights into the molecular characteristics of tumors, informing treatment decisions. For instance, elevated HER2 levels can indicate the appropriateness of therapies such as trastuzumab, specifically designed to target tumors that overexpress this protein [82].
Moreover, pharmacogenomics focuses on analyzing genetic variants, such as CYP2C19, to optimize drug dosing and selection for individual patients. Understanding how these genetic differences affect drug metabolism allows clinicians to customize treatments, minimizing adverse effects and enhancing efficacy. For example, insights from pharmacogenomics can help determine the appropriate dosage of 5-fluorouracil, a chemotherapy medication, thereby reducing the risk of toxicity. This personalized approach not only improves patient safety but also maximizes therapeutic benefits [83].
Finally, clustered regularly interspaced short palindromic repeats (CRISPR)-based gene editing represents a revolutionary technology that can correct cancer-related mutations or enhance immune cells for therapeutic purposes. This approach holds significant promise, especially in preclinical trials for solid tumors. By precisely targeting and modifying specific genetic sequences, researchers can repair the mutations driving cancer progression and boost the functionality of immune cells, allowing them to more effectively destroy tumor cells. Thus, the versatility of CRISPR technology enables innovative strategies in cancer treatment, potentially leading to more effective therapies that harness the body’s immune response to combat malignancies. By leveraging NGS and other molecular profiling techniques, oncologists can identify key genetic alterations that make cancer susceptible to targeted therapies [84,85].
Cancer’s “weak spots” refer to specific genetic, molecular, or biological vulnerabilities that treatments can exploit to disrupt tumor growth and survival. These vulnerabilities often stem from oncogenic driver mutations, such as EGFR in lung cancer or BRAF V600E in melanoma. These mutations play crucial roles in cancer progression and can be effectively inhibited by targeted therapies, such as osimertinib for EGFR mutations and vemurafenib for BRAF V600E mutations [86]. Dysregulated signaling pathways, such as the PI3K/AKT/mTOR pathway, present another critical avenue for intervention. Drugs like everolimus target these pathways to halt cell proliferation. Additionally, tumors rely on their microenvironment for essential resources and to evade the immune system. This reliance makes targets like vascular endothelial growth factor (VEGF), which is involved in anti-angiogenesis, and immune checkpoint inhibitors, such as pembrolizumab, effective strategies for treatment [87].
Synthetic lethality is another concept in cancer therapy that occurs when the simultaneous impairment of two genes leads to cell death, while the loss of either genos alone does not affect cell viability. This strategy exploits specific genetic vulnerabilities in tumor cells, particularly those with mutations in DNA repair genes like BRCA1 and BRCA2. In these cancers, the loss of BRCA function impairs the homologous recombination repair pathway, making cells reliant on alternative repair mechanisms. PARP inhibitors work by blocking the PARP enzyme, which is essential for repairing single-strand DNA breaks. When PARP is inhibited, the accumulation of unrepaired DNA damage leads to double-strand breaks that cannot be effectively repaired in BRCA-deficient cells, resulting in cell death. This approach enhances treatment efficacy while sparing normal cells, and ongoing research into other synthetic lethal interactions aims to broaden its application across various cancer types, offering promising avenues for personalized therapy [88]. Additionally, epigenetic alterations that modify gene expression without altering the DNA sequence offer opportunities for therapies aimed at reversing these changes. By identifying and leveraging these diverse weak spots, personalized molecular therapies can deliver precise and effective treatments tailored to each patient’s unique tumor biology [89,90].
Unlike traditional treatments, personalized therapies focus on the specific molecular quirks that drive tumor growth,, such as mutated genes, overactive proteins, or disrupted signaling pathways. The concept is both simple and revolutionary: every cancer has a unique weak spot, a flaw in its machinery that sustains its growth. By precisely targeting these vulnerabilities, it is possible to halt the disease without damaging surrounding healthy tissue. While traditional treatments often swing a wrecking ball, these therapies act with the precision of a scalpel, effectively dismantling cancer’s defenses and rewriting survival stories [86,91].
Personalized molecular therapies represent a transformative approach to cancer treatment by targeting specific vulnerabilities unique to each patient’s tumor (Figure 2). A cornerstone of these therapies is the use of targeted therapies, which block the molecular pathways essential for cancer cell survival. For example, tyrosine kinase inhibitors (TKIs) like imatinib target the BCR-ABL fusion protein in chronic myeloid leukemia, while trastuzumab inhibits HER2 in HER2-positive breast cancer, and vemurafenib blocks the BRAF mutation in melanoma [11,92].
Another key strategy involves immunotherapies, which leverage the immune system to combat cancer based on the tumor’s molecular profile. Immune checkpoint inhibitors, such as pembrolizumab, have proven effective in tumors with high PD-L1 expression or mismatch repair deficiency. Additionally, chimeric antigen receptor T cell (CAR-T cell) therapy involves engineering a patient’s T cells to specifically recognize and attack cancer antigens, enhancing the body’s ability to target and destroy malignant cells [93]. Furthermore, RNA-based therapies are emerging as a powerful tool, utilizing RNA interference (RNAi) or mRNA technologies to target cancer-driving genes. Small interfering RNAs (siRNAs) can silence oncogenic genes, while mRNA vaccines similar to those developed for COVID-19 are being explored to stimulate anti-cancer immune responses. Together, these approaches highlight the versatility and precision of personalized molecular therapies in combating cancer through tailored, mechanism-specific treatments [94,95].
Imagine a tumor as a factory operating out of control, relentlessly producing cells because a critical switch, such as a protein like EGFR or HER2, has become stuck in the “on” position. Molecular therapies target these faulty switches, either flipping them off or blocking them entirely, effectively cutting off the resources that cancer needs to grow. By disrupting these essential signals, these therapies can halt the relentless production of cancer cells and restore balance to the body [96]. This specificity spares healthy tissues, dramatically reducing the harsh side effects associated with traditional treatments. When combined with biomarker testing, this approach becomes even more effective. This is a shift from the ’carpet-bombing’ approach of conventional chemotherapy to the ’precision targeting’ of personalized medicine, and the results are proving that this targeted strategy is not just a scientific breakthrough but a life-changing reality for many patients [97,98].
Take chronic myeloid leukemia (CML) as an example, a disease that was once considered a death sentence. Today, it has been transformed into a manageable condition thanks to imatinib. This drug specifically targets the BCR-ABL protein that drives the cancer, enabling over 80% of patients to live for decades after diagnosis. This shift illustrates the power of targeted therapies in improving survival and quality of life for cancer patients [99]. Studies have shown that adding trastuzumab to standard care reduces the risk of recurrence by half, effectively transforming aggressive tumors into treatable conditions. This advancement highlights the critical role of targeted therapies in improving outcomes for patients with HER2-positive breast cancer [100].
Similarly, in melanoma, the BRAF mutation drives tumor growth in about half of cases, and vemurafenib effectively targets this mutation, rapidly shrinking tumors where chemotherapy often fails and transforming bleak prognoses into months or even years of extended life. These advances have demonstrated significant clinical impact, improving outcomes in real-world settings and providing patients with the chance to walk away from what were once considered fatal diagnoses [101,102].
This is where cancer treatment evolves from a blunt, one-size-fits-all approach to something sharper, smarter, and more humane (Table 4). Personalized therapies aim not just to destroy cancer but to outsmart it, using the tumor’s own biology as its critical vulnerability. While these treatments face challenges, such as the way in which some cancers develop resistance over time and the staggering costs that can reach tens of thousands of USD annually, the momentum behind this revolution is undeniable. These innovative approaches continue to reshape the landscape of cancer treatment, offering hope and improved outcomes for many patients [103].
Researchers are discovering new targets for treatment, like Kirsten rat sarcoma viral oncogene homolog (KRAS) in pancreatic cancer and Anaplastic Lymphoma Kinase (ALK) in lung cancer, and are improving combination therapies to stay ahead of resistance. The real strength of personalized medicine is demonstrated when these treatments are guided by biomarkers: simple blood tests that can identify the tumor’s weak spot, allowing the drug to make a precise attack. This gives us a glimpse of a future where cancer is not just a devastating diagnosis but a complex puzzle that we can solve step by step. This future offers patients not only more time but also a better quality of life. After years of using broad strategies, we are finally learning to focus our efforts and make the most effective impact where it matters [104].
Table 4. Cancer treatment: Conventional vs. advanced methods [105].
Table 4. Cancer treatment: Conventional vs. advanced methods [105].
AspectConventional TreatmentAdvanced Treatment
Techniques
  • Surgery (tumor removal)
  • Chemotherapy
  • Radiation therapy
  • Targeted therapy (EGFR inhibitors, BRAF inhibitors)
  • Immunotherapy (CAR-T cells, checkpoint inhibitors)
  • CRISPR gene editing
  • Personalized medicine (based on genetic profiling)
  • AI-driven drug discovery
SpecificityNon-specific, affecting both cancerous and healthy cellsHighly specific, targeting cancer cells while minimizing damage to normal cells
Side effectsHigh, including nausea, hair loss, immune suppressionReduced, as treatments are more focused and personalized
EffectivenessVariable, depends on cancer type and stageHigher efficacy in many cases, especially for resistant or rare cancers
Recurrence rateOften high due to incomplete tumor eradicationLower, as targeted treatments disrupt cancer pathways effectively
While traditional methods of cancer diagnosis and treatment remain essential, cutting-edge technologies are transforming the field by enabling earlier detection, greater precision, and more effective treatment interventions. These advancements empower healthcare providers to find cancers at their most treatable stages, significantly improving the likelihood of successful outcomes [106]. Currently, personalized medicine is becoming more precise by incorporating tools like genomic sequencing, molecular diagnostics, and AI-driven technologies into cancer care, tailoring therapies to each patient’s unique genetic profile and tumor biology. This targeted approach effectively addresses the underlying causes of cancer while minimizing damage to healthy cells, reducing side effects and enhancing patients’ quality of life. As these innovations evolve, they hold the potential to further improve cancer care, with AI and machine learning providing clinicians with real-time insights to guide smarter, data-driven decisions. The result is a new era of individualized cancer treatment, offering hope for better outcomes, improved survival rates, and a brighter future for those facing this challenging disease [107].

5. Bridging Cancer Diagnosis and Treatment: The Power of Integration for Better Health Care

The integration of cancer diagnosis and treatment marks a crucial shift in modern oncology, aiming to improve patient outcomes through a multifaceted approach. This seamless connection ensures that patients have a smoother journey from detection to recovery. Traditionally, delays between diagnosis and the start of treatment have been problematic, often due to logistical challenges, fragmented healthcare systems, and poor communication among specialists. However, cancer care is evolving. It is no longer just about identifying the disease and then choosing a treatment; instead, diagnosis and treatment now work together from the very beginning. By combining biomarkers with personalized molecular-targeted therapies, we create a powerful alliance that is changing the landscape of oncology and offering new hope for patients [108,109].
Early and accurate diagnosis is essential for effective cancer treatment, and advancements in diagnostic technologies are transforming how cancers are detected and characterized. For example, genomic profiling helps clinicians identify specific mutations driving a patient’s tumor, while liquid biopsies allow for the real-time monitoring of disease progression and treatment response through circulating tumor DNA. These innovations not only enable earlier detection but also provide valuable insights into the changing biology of a tumor, ensuring that treatment strategies can adapt and remain precise. When these diagnostic tools are effectively integrated with treatment options, they unlock the full potential of personalized medicine [110,111]. For instance, identifying actionable biomarkers through genomic testing can guide the selection of targeted therapies, such as tyrosine kinase inhibitors or immune checkpoint blockers, tailored to the genetic makeup of an individual’s cancer. Similarly, AI and machine learning algorithms can analyze vast datasets to predict treatment responses, optimize drug combinations, and anticipate resistance mechanisms, empowering clinicians to make informed decisions at every stage of care [112].
Biomarkers serve as detectives, accurately locating cancer through blood tests or tissue samples. At the same time, targeted therapies act like sharpshooters, hitting the tumor’s weak spots while protecting healthy cells from harm. When these two innovations work together, they create a powerful system where identifying the cancer leads directly to the most effective treatment plan. This tailored approach eliminates guesswork and focuses on the best solution. This integration represents not just a small improvement over traditional methods but a significant leap into a future where every aspect of cancer care, from detection to treatment, is carefully designed based on each patient’s unique disease biology. It is a bold move toward truly personalized medicine, offering hope for more efficient, effective, and compassionate care [5,104].
The true power of personalized cancer care comes from how biomarkers not only indicate the presence of cancer but also uncover its strategies. They reveal the specific mutations and pathways that are driving the disease [113]. For example, a simple blood test might reveal an ALK gene rearrangement in a lung cancer patient. Instead of using a trial-and-error approach with chemotherapy, doctors can prescribe crizotinib, a drug that specifically targets this mutation. The outcome? Tumors can shrink in weeks instead of months, reducing unnecessary suffering for patients [114]. Similarly, in colorectal cancer, if a biopsy reveals no KRAS mutations, it signals a green light for cetuximab, a therapy that targets EGFR, tripling response rates compared with standard treatments. Without this biomarker insight, the same therapy would be a shot in the dark, ineffective against cancers with KRAS mutations. This synergy between biomarkers and therapies cuts through the overwhelming diversity of cancer over 200 types, each with its own subtypes and transforms it into a manageable challenge. It is like having a global positioning system (GPS) for a maze: the biomarker points the way, and the therapy charts the route, dodging dead ends that waste time and hope [115,116].
The evidence supporting this approach is growing and extends beyond laboratory experiments; it is saving lives. In breast cancer, the MammaPrint test analyzes a 70-gene signature to predict the risk of recurrence. If the risk is low, patients can skip the harsh side effects of chemotherapy altogether. If the risk is high, doctors can customize treatment, such as combining trastuzumab with standard care for HER2-positive cases, which reduces the chances of relapse by 50% [117]. A groundbreaking trial in cervical cancer in 2022 took this a step further. By using tumor protein markers to guide a brief course of chemotherapy before standard treatment, researchers were able to reduce the risk of mortality by 40%. This life-saving achievement came from effectively matching the right drug to the right indicators [118]. Melanoma provides another clear example: PD-L1 biomarkers help identify patients who are likely to respond to pembrolizumab, an immunotherapy that boosts the immune system. For some patients, this means enjoying years of remission, while chemotherapy might have only offered months. Even rare cancers like gastrointestinal stromal tumors (GISTs) benefit from this strategy. When biomarkers detect KIT mutations, imatinib can be used, increasing the survival rate from a dismal 10% to over 80%. These effective pairings are not just coincidences; they demonstrate that connecting diagnosis to treatment is not only smart but also transformative [119,120].
The integration of biomarkers and targeted therapies is not a fixed process; it is a dynamic and evolving approach that adjusts as cancer changes. Researchers are increasingly using combination treatments, such as pairing targeted drugs with immunotherapy or radiation. This is guided by multi-marker panels that provide a comprehensive view of a tumor’s biology, including its DNA, proteins, and microenvironment. For example, in lung cancer, patients with EGFR mutations might begin treatment with osimertinib. If resistance develops, a circulating tumor DNA (ctDNA) test can identify new mutations like T790M, allowing for a switch to a next-generation drug that is tailored to the tumor’s evolving characteristics [5,121]. This adaptive process transforms cancer care into a responsive and personalized strategy. It reduces reliance on toxic treatments, leading to shorter hospital stays, less strain on patients, and greater potential for use in resource-limited settings. However, challenges, such as high costs, limited access to advanced diagnostics, and gaps in training for healthcare providers, mean this approach is not yet widely available. Despite these obstacles, the vision remains clear: a future where a cancer diagnosis not only identifies the disease but also provides the tools to fight it with precision, offering unprecedented control over what was once an overwhelming adversary [103,122].
To overcome obstacles in cancer treatment and diagnosis, a multifaceted approach is needed. Reducing costs through policy changes and promoting affordable treatment options can help alleviate financial barriers. Expanding access to advanced diagnostics, particularly in underserved areas, and utilizing telemedicine for remote consultations can improve early detection. Additionally, enhancing education and training for healthcare providers on the latest therapies ensures better patient care. Supporting research initiatives fosters innovation in treatment and diagnostics, while collaboration between public and private sectors can optimize resources. Together, these strategies can enhance accessibility and effectiveness in cancer care [123].
When diagnosis and treatment are aligned, healthcare systems can reduce redundancies, optimize workflows, and allocate resources more efficiently, ultimately lowering costs and improving accessibility. For patients, this approach creates a cohesive and supportive care journey, addressing their unique needs holistically from early detection and risk assessment to personalized therapies and survivorship planning. This unified strategy not only enhances survival rates but also improves the overall patient experience, paving the way for a future where cancer care is precise, personalized, and profoundly effective [124,125].
Looking ahead, the ongoing improvement of integrated cancer care holds great promise. New technologies, like single-cell sequencing, CRISPR-based diagnostics, and nanotechnology for drug delivery, are making it easier to combine diagnosis and treatment into a smooth process. This teamwork not only boosts our ability to fight cancer but also changes the way we think about compassionate, patient-centered healthcare. Connecting diagnosis and treatment is more than just a scientific breakthrough; it represents a major shift toward better health outcomes and a higher quality of life for patients. By encouraging collaboration across different fields and using the latest technologies, we are creating a future where cancer care is proactive, predictive, and truly personalized [126,127].

6. In Cancer: Real-World Wins and Challenges Ahead

The fight against cancer has made significant strides, largely due to the emergence of personalized medicine and targeted therapies. For many years, cancer research felt like a relentless race against time, but now the finish line appears nearer than ever. Real-world successes achieved through biomarkers and tailored treatments are transforming lives, turning once grim diagnoses into narratives of resilience and hope. These breakthroughs illustrate that combining advanced detection methods with customized treatment plans can effectively counteract cancer’s elusive strategies [103]. Patients are experiencing fewer side effects from chemotherapy or even avoiding it altogether, while others are surpassing survival expectations once thought unattainable. Each success is backed by hard data and human stories, proving just how far science has come. Yet, for every step forward, challenges remain; high costs, limited access, and unforeseen setbacks remind us that this revolution is far from complete. This section explores what is working, who is benefiting, and the obstacles still standing in the way, offering a clear-eyed view of both progress and the road ahead [128,129].
The successes in cancer diagnosis and treatment are undeniable. Take melanoma, which was once viewed as a relentless killer and is now responding to therapies like pembrolizumab. Patients with PD-L1 biomarkers have experienced significant tumor shrinkage, with some achieving five-year survival milestones that seemed unimaginable just a decade ago, when chemotherapy provided only a few extra months of life [130]. In lung cancer, drugs like osimertinib are doubling progression-free survival for patients with EGFR mutations, extending stable health to nearly two years compared with less than one with older treatments [131]. Breast cancer patients with low-risk MammaPrint scores are now able to forgo chemotherapy altogether, thriving instead on lighter therapies. Meanwhile, HER2-positive cases are benefiting from trastuzumab, which has halved recurrence rates. Even rare cancers, like gastrointestinal stromal tumors (GIST), are experiencing remarkable outcomes: imatinib effectively targets KIT mutations, boosting survival rates to 80%, a staggering increase from just 10% in the past. These successes are not isolated; global statistics support this encouraging trend [132,133].
According to the American Cancer Society, targeted therapies have significantly increased five-year survival rates for various cancer types by 10–20% since 2010. These statistics represent profound advancements in cancer treatment that are changing lives. Behind these numbers are real individuals, such as a 60-year-old teacher in Texas. After being diagnosed with lung cancer, she underwent an ALK test that identified her as a suitable candidate for crizotinib. The results were remarkable; her cancer responded dramatically to the treatment, allowing her to regain her health and continue her teaching career. Stories like hers underscore that this progress is not merely about data; it represents real deliverance for patients and their families, offering hope and renewed possibilities in the face of a daunting diagnosis. Each success story reflects the transformative power of personalized medicine and the impact it has on the lives of those it touches [134,135].
This progress reflects not only scientific creativity but also the profound impact of precision medicine. By understanding the unique genetic and molecular profiles of individual cancers, researchers and clinicians are developing treatments that work smarter, not harder. However, as transformative as these advancements are, they also reveal significant disparities in access and affordability, raising concerns about whether the benefits will reach everyone equally. While the strides made thus far inspire optimism and hope, it is crucial to address these barriers to ensure that the fight against cancer becomes a victory for all. Ensuring equitable access to these innovative therapies will be essential in making sure that no one is left behind in this battle, allowing every patient the opportunity for a better outcome and a chance at life [136].
Despite the remarkable progress in cancer diagnosis and treatment, not everyone benefits equally, and this is where the challenges become most evident. Resistance to targeted therapies continues to pose a significant obstacle, as cancers can evolve and develop new mutations that enable them to evade even the most advanced treatments. Additionally, financial barriers exacerbate the issue: these life-saving therapies often come with astronomical costs. For instance, the cost of osimertinib can exceed 15,000 USD per month, and insurance coverage is far from guaranteed. This leaves patients in wealthier nations grappling with affordability, while those in low-income regions face even greater challenges in accessing necessary care. Addressing these inequities is crucial to ensure that the advancements in cancer treatment benefit everyone, regardless of their financial situation [137]. Access is an even greater challenge; 70% of cancer deaths occur in places like sub-Saharan Africa and rural Asia, where access to biomarker testing and targeted drugs is severely limited. Imagine a farmer in Kenya waiting months or even a year for a basic diagnostic scan, let alone cutting-edge tools like ctDNA panels. The COVID-19 pandemic further exacerbated these disparities, disrupting screening programs and creating significant backlogs. According to the World Health Organization (WHO), early detection rates dropped by 30% in 2020 alone, giving cancer a dangerous head start [138,139].
Resistance to treatments adds another layer of complexity in the fight against cancer. Tumors can adapt rapidly, rendering drugs like vemurafenib ineffective within just a few months, which in turn forces researchers to race against time to develop next-generation solutions [140]. For instance, resistance to EGFR inhibitors in lung cancer often occurs due to secondary mutations such as T790M, highlighting the need for newer, more sophisticated drugs. However, tackling resistance requires more than just innovation; it necessitates systemic changes across the healthcare landscape. Doctors need specialized training, laboratories must be equipped with advanced technology, and healthcare systems need adequate funding to bridge gaps in care delivery. Even when there is a strong desire to implement these changes, resource constraints can significantly slow the rollout of breakthroughs. High costs and unequal access to cutting-edge therapies perpetuate stark disparities in care, especially in low-resource settings. Moreover, the complexity of diagnosing and treating cancer continues to increase, underscoring the urgent need for ongoing innovation. Tools like liquid biopsies and AI-driven analytics hold immense promise for staying ahead of the disease, but they require substantial investments in research, infrastructure, and training. Ensuring that these advancements reach all patients will be crucial in the collective fight against cancer [141,142].
Looking ahead, the key to progress lies in addressing these challenges while building on the successes already achieved. These obstacles are not impossible to overcome; they represent the next frontier in the fight against cancer. The victories we have seen thus far demonstrate what is possible: lives extended, suffering reduced, and hope restored. Bridging the gaps requires scaling up effective strategies, such as developing more affordable diagnostic tests, expanding access to therapies, and combining treatments in innovative ways to improve outcomes for patients worldwide. While the journey is complex and uneven, every patient thriving today stands as a beacon of hope for the future. For instance, combining therapies such as pairing targeted drugs with immunotherapy or radiation offers promising opportunities to outsmart resistant tumors. Biomarkers and targeted therapies are fundamentally rewriting cancer’s narrative, transforming it from one of despair into one of possibility. Now, the mission is clear: we must ensure that this transformative progress reaches every corner of the globe, leaving no patient behind. This commitment to equity in cancer care will be essential in turning hope into reality for all those affected by this disease [104,143].

7. The Road Forward: Scaling the Revolution Against Cancer

The road forward in the fight against cancer is defined by innovation, collaboration, and a strong determination to transform how we approach this disease on a global scale. The next chapter in cancer care is not just about refining existing tools; it is about reimagining how we prevent, detect, and treat cancer to create a future where its burden is significantly reduced. At the heart of this transformation are emerging technologies like artificial intelligence (AI) and liquid biopsies, which together have the potential to shift the focus from reactive treatment to proactive prevention. By harnessing these advancements, we can pave the way for earlier diagnoses and more effective interventions, ultimately improving outcomes for patients everywhere [144,145].
Artificial intelligence (AI) is revolutionizing cancer diagnostics by significantly enhancing both the speed and accuracy of clinical assessments. Utilizing deep learning and machine learning algorithms, AI can process and interpret vast datasets, including radiologic images, pathology slides, and electronic health records, with a level of precision that often surpasses human capabilities. For instance, in the case of lung cancer detection, convolutional neural networks (CNNs) can identify subtle nodular patterns and textural changes in CT scans that are indicative of early-stage malignancies, sometimes before they are visible to the human eye. Moreover, AI-powered platforms can integrate multi-omics data—such as genomic, transcriptomic, and proteomic profiles—to uncover molecular biomarkers like EGFR mutations or PD-L1 expression, which guide the selection of targeted therapies and immunotherapies. Clinically, this allows oncologists to move beyond a “one-size-fits-all” approach, tailoring treatment plans based on tumor biology and patient-specific factors. As AI continues to be embedded into diagnostic workflows, it not only improves early detection and prognostic accuracy but also facilitates real-time decision-making in precision oncology, ultimately improving patient outcomes and resource efficiency in healthcare systems [145].
At the same time, liquid biopsies are transforming the landscape of early cancer detection by providing a minimally invasive method by which to monitor tumor dynamics over time. Unlike traditional tissue biopsies, which are often limited by accessibility and invasiveness, liquid biopsies analyze circulating tumor DNA (ctDNA) and other tumor-derived biomarkers from a simple blood draw. This approach enables clinicians to detect molecular alterations associated with early-stage malignancies or tumor recurrence, often months before abnormalities become visible through imaging techniques like CT or MRI. Mechanistically, ctDNA reflects real-time genomic changes in tumor cells, offering a dynamic snapshot of tumor burden, mutation profiles, and potential resistance mechanisms. Companies such as Guardant Health have pioneered the clinical implementation of this technology, demonstrating its effectiveness in identifying high-risk cancers such as pancreatic and lung cancers at an earlier, more treatable stage. By facilitating prompt and personalized therapeutic intervention, liquid biopsies not only enhance clinical decision-making but also hold the potential to significantly improve progression-free survival and overall patient outcomes across a range of malignancies [146,147].
However, the true revolution lies in scaling these innovations so that they extend far beyond elite research centers and into communities worldwide. Prevention will play a pivotal role in this vision. Imagine AI-driven risk models that identify individuals with inherited mutations, such as BRCA1, for closer monitoring, or population-wide screening programs using liquid biopsies to detect aggressive cancers before they metastasize. Democratizing access to these tools is equally critical. Initiatives like the World Health Organization’s push for affordable diagnostics in low-income countries, along with partnerships between biotech firms and governments to subsidize AI-powered pathology tools, exemplify efforts to make cutting-edge technologies accessible to all. For instance, PathAI’s work in streamlining tumor analysis could soon be adapted for rural clinics, reducing costs and accelerating diagnoses in areas where specialists are scarce. By ensuring that everyone has access to these advancements, we can make significant strides in cancer prevention and treatment on a global scale [148,149].
This forward-looking vision has the potential to dramatically reduce cancer’s toll. By catching diseases earlier, tailoring treatments more precisely, and ensuring that no patient is left behind due to geography or income, these advancements could significantly lower the global mortality rate, which currently stands at 10 million annually, according to the International Agency for Research on Cancer. Moreover, easing the economic strain on healthcare systems would create ripple effects, enhancing the quality of life for millions. By prioritizing equitable access to innovative technologies and treatments, we can create a healthier future for everyone, regardless of their circumstances [26].
Generally, “The Road Forward” in the fight against cancer is not just a hopeful slogan; it is a call to action. It urges scientists, policymakers, and innovators to connect today’s breakthroughs with tomorrow’s reality, transforming cancer into a manageable chapter in human health rather than a defining one. Through collaboration and commitment, we can turn the promise of these innovations into tangible progress, ensuring that the fight against cancer becomes a victory for everyone, everywhere. By working together, we can create a future where effective prevention, early detection, and personalized treatment are accessible to all, ultimately improving lives across the globe [150].

8. Conclusions: A New Era in Cancer Care

Cancer care is undergoing a revolutionary transformation, driven by the integration of biomarker-based diagnosis and personalized molecular-targeted treatments. Traditional approaches like chemotherapy and radiation, while effective, often impose significant burdens on patients due to their non-specific nature, impacting both malignant and healthy cells. However, advancements in genomics and molecular diagnostics have ushered in the era of precision medicine, enabling oncologists to provide therapies that are not only more effective but also significantly less toxic. This shift marks a pivotal moment in cancer treatment, offering hope for better outcomes and improved quality of life for patients worldwide [50,151].
As we navigate this exciting landscape, it is crucial to reflect on the ongoing challenges in the application of biomarkers and targeted molecular treatments. One significant issue is the heterogeneity of tumors, which can lead to variable responses to targeted therapies. While biomarkers have revolutionized our approach to cancer treatment, their effectiveness can be hampered by the presence of multiple genetic alterations within a single tumor. This complexity necessitates a deeper understanding of the tumor microenvironment and its influence on treatment outcomes [152].
Moreover, there remains a pressing need for comprehensive biomarker testing to ensure that all patients receive the most appropriate therapies. Access to these advanced diagnostic tools is not uniform, leading to disparities in treatment efficacy among different populations. Additionally, as targeted therapies become more prevalent, the challenge of acquired resistance becomes increasingly important. Understanding the mechanisms that lead to resistance will be critical for developing next-generation treatments [153].
Looking ahead, several emerging research directions warrant attention. First, the integration of artificial intelligence (AI) and multi-omics data should be prioritized to refine biomarker discovery and optimize therapeutic strategies. AI-driven predictive models can analyze vast genomic datasets, accelerating drug development and enabling more precise treatment personalization. Furthermore, the convergence of biomarker-based diagnosis with innovative gene-editing technologies like CRISPR holds immense promise for unlocking new frontiers in targeted therapy [153,154]. Another priority should be the exploration of novel therapeutic combinations that address resistance mechanisms. Research focusing on the tumor microenvironment and its influence on treatment efficacy could lead to breakthroughs in managing advanced cancers. Additionally, expanding access to genomic testing and personalized treatments across diverse patient populations is essential to ensure equitable cancer care [155].
In conclusion, the future of cancer care depends on our ability to understand the complexities of the disease and address ongoing challenges. By enhancing access to comprehensive biomarker testing, we can ensure that all patients receive personalized and effective treatments, reducing disparities in care [52].
Investing in research to understand resistance mechanisms will be crucial for developing new therapies, while leveraging artificial intelligence and multi-omics data can accelerate drug development and improve treatment personalization. Additionally, exploring combination therapies may enhance effectiveness, and fostering collaboration among researchers, clinicians, and patients will help translate scientific discoveries into practical solutions. Together, these efforts represent not just a commitment to advancing cancer care but the beginning of a new era, one in which innovative approaches lead to more effective and hopeful treatment options for patients [156,157].

Author Contributions

G.M. and M.B. conceptualized and wrote the manuscript. All authors contributed to the article and approved the submitted version. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AFPAlpha-fetoprotein
AI Artificial intelligence
AKT Protein kinase B
ALK Anaplastic lymphoma kinase
BRCA Breast cancer gene
CA-125Cancer antigen 125
CAR-T Chimeric antigen receptor T cell
CDT Carbohydrate-deficient transferrin
CEACarcinoembryonic antigen
CML Chronic myelogenous leukemia
CNNsConvolutional neural networks
CRISPR Clustered regularly interspaced short palindromic repeats
CT scansComputed tomography scans
CTCsCirculating tumor cells
ctDNA Circulating tumor DNA
CUP Cancer of unknown primary
DNA Deoxyribonucleic acid
DX Diagnosis
EGFR Epidermal growth factor receptor
GIST Gastrointestinal stromal tumor
GPS Global positioning system
HCGHuman chorionic gonadotropin
HER2Human epidermal growth factor receptor 2
HPVHuman papillomavirus
KIT V-Kit Hardy–Zuckerman 4 feline sarcoma viral oncogene homolog
KRAS Kirsten rat sarcoma viral oncogene homolog
LDHLactate dehydrogenase
MRIMagnetic resonance imaging
mRNA Messenger ribonucleic acid
MSI-HMicrosatellite instability-high
mTOR Mechanistic target of rapamycin
NGS Next-generation sequencing
NMP22Nuclear matrix protein 22
PD-L1 Programmed death-ligand 1
PI3K Phosphoinositide 3-kinase
PSA Prostate-specific antigen
RNA Ribonucleic acid
RNAi RNA interference
scRNA-seqssngle-cell RNA sequencing
siRNAssmall interfering RNAs
T790MRefers to a mutation in the epidermal growth factor receptor (EGFR) gene
TKI Tyrosine kinase inhibitor
TP53Tumor protein p53
VEGF Vascular endothelial growth factor
WES Whole exome sequencing

References

  1. Carneiro, B.A.; El-Deiry, W.S. Targeting apoptosis in cancer therapy. Nat. Rev. Clin. Oncol. 2020, 17, 395–417. [Google Scholar] [CrossRef]
  2. Carrillo-Beltrán, D.; Osorio, J.C.; Blanco, R.; Oliva, C.; Boccardo, E.; Aguayo, F. Interaction between cigarette smoke and human papillomavirus 16 E6/E7 oncoproteins to induce SOD2 expression and DNA damage in head and neck cancer. Int. J. Mol. Sci. 2023, 24, 6907. [Google Scholar] [CrossRef] [PubMed]
  3. Tigu, A.B.; Tomuleasa, C. Exploring Novel Frontiers in Cancer Therapy. Biomedicines 2024, 12, 1345. [Google Scholar] [CrossRef] [PubMed]
  4. Huerta, E.; Grey, N. Cancer Control Opportunities in Low- and Middle-Income Countries; National Academies Press: Washington, DC, USA, 2007; Volume 57, pp. 72–74. [Google Scholar]
  5. Nakamura, Y.; Kawazoe, A.; Lordick, F.; Janjigian, Y.Y.; Shitara, K. Biomarker-targeted therapies for advanced-stage gastric and gastro-oesophageal junction cancers: An emerging paradigm. Nat. Rev. Clin. Oncol. 2021, 18, 473–487. [Google Scholar] [CrossRef]
  6. Buzdin, A.; Sorokin, M.; Garazha, A.; Sekacheva, M.; Kim, E.; Zhukov, N.; Wang, Y.; Li, X.; Kar, S.; Hartmann, C. Molecular pathway activation–new type of biomarkers for tumor morphology and personalized selection of target drugs. Semin. Cancer biology 2018, 53, 110–124. [Google Scholar] [CrossRef] [PubMed]
  7. Pashayan, N.; Pharoah, P.D. The challenge of early detection in cancer. Science 2020, 368, 589–590. [Google Scholar] [CrossRef] [PubMed]
  8. Huang, Z.; Ma, L.; Huang, C.; Li, Q.; Nice, E.C. Proteomic profiling of human plasma for cancer biomarker discovery. Proteomics 2017, 17, 1600240. [Google Scholar] [CrossRef]
  9. 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]
  10. Sicklick, J.K.; Kato, S.; Okamura, R.; Schwaederle, M.; Hahn, M.E.; Williams, C.B.; De, P.; Krie, A.; Piccioni, D.E.; Miller, V.A. Molecular profiling of cancer patients enables personalized combination therapy: The I-PREDICT study. Nat. Med. 2019, 25, 744–750. [Google Scholar] [CrossRef]
  11. Kim, K.B. Personalized therapy in oncology: Melanoma as a paradigm for molecular-targeted treatment approaches. Clin. Exp. Metastasis 2024, 41, 465–471. [Google Scholar] [CrossRef]
  12. Nami, B.; Maadi, H.; Wang, Z. Mechanisms underlying the action and synergism of trastuzumab and pertuzumab in targeting HER2-positive breast cancer. Cancers 2018, 10, 342. [Google Scholar] [CrossRef] [PubMed]
  13. Kunte, S.; Abraham, J.; Montero, A.J. Novel HER2–targeted therapies for HER2–positive metastatic breast cancer. Cancer 2020, 126, 4278–4288. [Google Scholar] [CrossRef] [PubMed]
  14. Ke, X.; Shen, L. Molecular targeted therapy of cancer: The progress and future prospect. Front. Lab. Med. 2017, 1, 69–75. [Google Scholar] [CrossRef]
  15. Nasser, A.; Hussein, A.; Chamba, C.; Yonazi, M.; Mushi, R.; Schuh, A.; Luzzatto, L. Molecular response to imatinib in patients with chronic myeloid leukemia in Tanzania. Blood Adv. 2021, 5, 1403–1411. [Google Scholar] [CrossRef]
  16. Wu, J.; Lin, Z. Non-small cell lung cancer targeted therapy: Drugs and mechanisms of drug resistance. Int. J. Mol. Sci. 2022, 23, 15056. [Google Scholar] [CrossRef]
  17. Hochhaus, A.; Larson, R.A.; Guilhot, F.; Radich, J.P.; Branford, S.; Hughes, T.P.; Baccarani, M.; Deininger, M.W.; Cervantes, F.; Fujihara, S. Long-term outcomes of imatinib treatment for chronic myeloid leukemia. N. Engl. J. Med. 2017, 376, 917–927. [Google Scholar] [CrossRef]
  18. Tenchov, R.; Sapra, A.K.; Sasso, J.; Ralhan, K.; Tummala, A.; Azoulay, N.; Zhou, Q.A. Biomarkers for early cancer detection: A landscape view of recent advancements, spotlighting pancreatic and liver cancers. ACS Pharmacol. Transl. Sci. 2024, 7, 586–613. [Google Scholar] [CrossRef] [PubMed]
  19. Han, X.; Wang, J.; Sun, Y. Circulating tumor DNA as biomarkers for cancer detection. Genom. Proteom. Bioinform. 2017, 15, 59–72. [Google Scholar] [CrossRef]
  20. Hunter, B.; Hindocha, S.; Lee, R.W. The role of artificial intelligence in early cancer diagnosis. Cancers 2022, 14, 1524. [Google Scholar] [CrossRef]
  21. Mahmood, H.; Shaban, M.; Rajpoot, N.; Khurram, S.A. Artificial Intelligence-based methods in head and neck cancer diagnosis: An overview. Br. J. Cancer 2021, 124, 1934–1940. [Google Scholar] [CrossRef]
  22. Dessale, M.; Mengistu, G.; Mengist, H.M. Nanotechnology: A promising approach for cancer diagnosis, therapeutics and theragnosis. Int. J. Nanomed. 2022, 17, 3735. [Google Scholar] [CrossRef] [PubMed]
  23. Schaffer, A.L.; Pearson, S.-A.; Perez-Concha, O.; Dobbins, T.; Ward, R.L.; van Leeuwen, M.T.; Rhee, J.J.; Laaksonen, M.A.; Craigen, G.; Vajdic, C.M. Diagnostic and health service pathways to diagnosis of cancer-registry notified cancer of unknown primary site (CUP). PLoS ONE 2020, 15, e0230373. [Google Scholar] [CrossRef] [PubMed]
  24. Pauli, C.; Bochtler, T.; Mileshkin, L.; Baciarello, G.; Losa, F.; Ross, J.S.; Pentheroudakis, G.; Zarkavelis, G.; Yalcin, S.; Özgüroğlu, M. A challenging task: Identifying patients with cancer of unknown primary (CUP) according to ESMO guidelines: The CUPISCO trial experience. Oncologist 2021, 26, e769–e779. [Google Scholar] [CrossRef]
  25. Mathew, B.G.; Aliyuda, F.; Taiwo, D.; Adekeye, K.; Agada, G.; Sanchez, E.; Ghose, A.; Rassy, E.; Boussios, S. From biology to diagnosis and treatment: The Ariadne’s thread in cancer of unknown primary. Int. J. Mol. Sci. 2023, 24, 5588. [Google Scholar] [CrossRef] [PubMed]
  26. Crosby, D.; Bhatia, S.; Brindle, K.M.; Coussens, L.M.; Dive, C.; Emberton, M.; Esener, S.; Fitzgerald, R.C.; Gambhir, S.S.; Kuhn, P. Early detection of cancer. Science 2022, 375, eaay9040. [Google Scholar] [CrossRef]
  27. Gillies, R.J.; Schabath, M.B. Radiomics improves cancer screening and early detection. Cancer Epidemiol. Biomark. Prev. 2020, 29, 2556–2567. [Google Scholar] [CrossRef]
  28. Anghel, S.A.; Ioniță-Mîndrican, C.-B.; Luca, I.; Pop, A.L. Promising epigenetic biomarkers for the early detection of colorectal cancer: A systematic review. Cancers 2021, 13, 4965. [Google Scholar] [CrossRef]
  29. Wu, L.; Qu, X. Cancer biomarker detection: Recent achievements and challenges. Chem. Soc. Rev. 2015, 44, 2963–2997. [Google Scholar] [CrossRef]
  30. Haga, Y.; Uemura, M.; Baba, S.; Inamura, K.; Takeuchi, K.; Nonomura, N.; Ueda, K. Identification of multisialylated LacdiNAc structures as highly prostate cancer specific glycan signatures on PSA. Anal. Chem. 2019, 91, 2247–2254. [Google Scholar] [CrossRef]
  31. Ebell, M.H.; Culp, M.B.; Radke, T.J. A systematic review of symptoms for the diagnosis of ovarian cancer. Am. J. Prev. Med. 2016, 50, 384–394. [Google Scholar] [CrossRef]
  32. Orr, B.; Edwards, R.P. Diagnosis and treatment of ovarian cancer. Hematol./Oncol. Clin. 2018, 32, 943–964. [Google Scholar] [CrossRef] [PubMed]
  33. Young Bang, J.; Krall, K.; Jhala, N.; Singh, C.; Tejani, M.; Arnoletti, J.P.; Navaneethan, U.; Hawes, R.; Varadarajulu, S. Comparing Needles and Methods of Endoscopic Ultrasound-Guided Fine-Needle Biopsy to Optimize Specimen Quality and Diagnostic Accuracy for Patients With Pancreatic Masses in a Randomized Trial. Clin. Gastroenterol. Hepatol. 2021, 19, 825–835.e7. [Google Scholar] [CrossRef]
  34. Williamson, J.M.; Hocken, D.B. Pancreatic cancer in the media: The Swayze shift. Ann. R. Coll. Surg. Engl. 2010, 92, 537–538. [Google Scholar] [CrossRef][Green Version]
  35. Chase, D.M.; Neighbors, J.; Perhanidis, J.; Monk, B.J. Gastrointestinal symptoms and diagnosis preceding ovarian cancer diagnosis: Effects on treatment allocation and potential diagnostic delay. Gynecol. Oncol. 2021, 161, 832–837. [Google Scholar] [CrossRef]
  36. Dilley, J.; Burnell, M.; Gentry-Maharaj, A.; Ryan, A.; Neophytou, C.; Apostolidou, S.; Karpinskyj, C.; Kalsi, J.; Mould, T.; Woolas, R.; et al. Ovarian cancer symptoms, routes to diagnosis and survival—Population cohort study in the ‘no screen’ arm of the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS). Gynecol. Oncol. 2020, 158, 316–322. [Google Scholar] [CrossRef] [PubMed]
  37. Wang, X.; Zhang, H.; Chen, X. Drug resistance and combating drug resistance in cancer. Cancer Drug Resist. 2019, 2, 141–160. [Google Scholar] [CrossRef]
  38. Chatterjee, N.; Bivona, T.G. Polytherapy and Targeted Cancer Drug Resistance. Trends Cancer 2019, 5, 170–182. [Google Scholar] [CrossRef]
  39. Cao, C.; Wang, D.; Chung, C.; Tian, D.; Rimner, A.; Huang, J.; Jones, D.R. A systematic review and meta-analysis of stereotactic body radiation therapy versus surgery for patients with non-small cell lung cancer. J. Thorac. Cardiovasc. Surg. 2019, 157, 362–373.e8. [Google Scholar] [CrossRef] [PubMed]
  40. Cheng, B.; He, H.; Chen, B.; Zhou, Q.; Luo, T.; Li, K.; Du, T.; Huang, H. Assessment of treatment outcomes: Cytoreductive surgery compared to radiotherapy in oligometastatic prostate cancer—An in-depth quantitative evaluation and retrospective cohort analysis. Int. J. Surg. 2024, 110, 3190–3202. [Google Scholar] [CrossRef]
  41. Camerini, A.; Mazzoni, F.; Scotti, V.; Tibaldi, C.; Sbrana, A.; Calabrò, 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]
  42. Wan, Y.; Wang, J.; Xu, J.F.; Tang, F.; Chen, L.; Tan, Y.Z.; Rao, C.L.; Ao, H.; Peng, C. Panax ginseng and its ginsenosides: Potential candidates for the prevention and treatment of chemotherapy-induced side effects. J. Ginseng Res. 2021, 45, 617–630. [Google Scholar] [CrossRef] [PubMed]
  43. Mukherjee, S. Genomics-Guided Immunotherapy for Precision Medicine in Cancer. Cancer Biother. Radiopharm. 2019, 34, 487–497. [Google Scholar] [CrossRef] [PubMed]
  44. Scheetz, L.; Park, K.S.; Li, Q.; Lowenstein, P.R.; Castro, M.G.; Schwendeman, A.; Moon, J.J. Engineering patient-specific cancer immunotherapies. Nat. Biomed. Eng. 2019, 3, 768–782. [Google Scholar] [CrossRef]
  45. Pereira, S.P.; Oldfield, L.; Ney, A.; Hart, P.A.; Keane, M.G.; Pandol, S.J.; Li, D.; Greenhalf, W.; Jeon, C.Y.; Koay, E.J.; et al. Early detection of pancreatic cancer. Lancet Gastroenterol. Hepatol. 2020, 5, 698–710. [Google Scholar] [CrossRef] [PubMed]
  46. Farr, K.P.; Moses, D.; Haghighi, K.S.; Phillips, P.A.; Hillenbrand, C.M.; Chua, B.H. Imaging Modalities for Early Detection of Pancreatic Cancer: Current State and Future Research Opportunities. Cancers 2022, 14, 2539. [Google Scholar] [CrossRef]
  47. Chinnappan, R.; Mir, T.A.; Alsalameh, S.; Makhzoum, T.; Alzhrani, A.; Alnajjar, K.; Adeeb, S.; Al Eman, N.; Ahmed, Z.; Shakir, I.; et al. Emerging Biosensing Methods to Monitor Lung Cancer Biomarkers in Biological Samples: A Comprehensive Review. Cancers 2023, 15, 3414. [Google Scholar] [CrossRef]
  48. Tsimberidou, A.M.; Fountzilas, E.; Nikanjam, M.; Kurzrock, R. Review of precision cancer medicine: Evolution of the treatment paradigm. Cancer Treat. Rev. 2020, 86, 102019. [Google Scholar] [CrossRef]
  49. Kwong, G.A.; Ghosh, S.; Gamboa, L.; Patriotis, C.; Srivastava, S.; Bhatia, S.N. Synthetic biomarkers: A twenty-first century path to early cancer detection. Nat. Rev. Cancer 2021, 21, 655–668. [Google Scholar] [CrossRef]
  50. Kalia, M. Biomarkers for personalized oncology: Recent advances and future challenges. Metabolism 2015, 64, S16–S21. [Google Scholar] [CrossRef]
  51. Zakari, S.; Niels, N.K.; Olagunju, G.V.; Nnaji, P.C.; Ogunniyi, O.; Tebamifor, M.; Israel, E.N.; Atawodi, S.E.; Ogunlana, O.O. Emerging biomarkers for non-invasive diagnosis and treatment of cancer: A systematic review. Front. Oncol. 2024, 14, 1405267. [Google Scholar] [CrossRef]
  52. Sarhadi, V.K.; Armengol, G. Molecular Biomarkers in Cancer. Biomolecules 2022, 12, 1021. [Google Scholar] [CrossRef] [PubMed]
  53. Gautam, S.K.; Khan, P.; Natarajan, G.; Atri, P.; Aithal, A.; Ganti, A.K.; Batra, S.K.; Nasser, M.W.; Jain, M. Mucins as Potential Biomarkers for Early Detection of Cancer. Cancers 2023, 15, 1640. [Google Scholar] [CrossRef] [PubMed]
  54. Li, J.; Guan, X.; Fan, Z.; Ching, L.M.; Li, Y.; Wang, X.; Cao, W.M.; Liu, D.X. Non-Invasive Biomarkers for Early Detection of Breast Cancer. Cancers 2020, 12, 2767. [Google Scholar] [CrossRef] [PubMed]
  55. Mohamed, E.; García Martínez, D.J.; Hosseini, M.S.; Yoong, S.Q.; Fletcher, D.; Hart, S.; Guinn, B.A. Identification of biomarkers for the early detection of non-small cell lung cancer: A systematic review and meta-analysis. Carcinogenesis 2024, 45, 1–22. [Google Scholar] [CrossRef]
  56. Nicolini, A.; Ferrari, P.; Duffy, M.J. Prognostic and predictive biomarkers in breast cancer: Past, present and future. Semin. Cancer Biol. 2018, 52, 56–73. [Google Scholar] [CrossRef]
  57. Das, V.; Kalita, J.; Pal, M. Predictive and prognostic biomarkers in colorectal cancer: A systematic review of recent advances and challenges. Biomed. Pharmacother. 2017, 87, 8–19. [Google Scholar] [CrossRef]
  58. Pabst, L.; Lopes, S.; Bertrand, B.; Creusot, Q.; Kotovskaya, M.; Pencreach, E.; Beau-Faller, M.; Mascaux, C. Prognostic and Predictive Biomarkers in the Era of Immunotherapy for Lung Cancer. Int. J. Mol. Sci. 2023, 24, 7577. [Google Scholar] [CrossRef]
  59. Duffy, M.J.; Harbeck, N.; Nap, M.; Molina, R.; Nicolini, A.; Senkus, E.; Cardoso, F. Clinical use of biomarkers in breast cancer: Updated guidelines from the European Group on Tumor Markers (EGTM). Eur. J. Cancer 2017, 75, 284–298. [Google Scholar] [CrossRef]
  60. Harris, L.N.; Ismaila, N.; McShane, L.M.; Andre, F.; Collyar, D.E.; Gonzalez-Angulo, A.M.; Hammond, E.H.; Kuderer, N.M.; Liu, M.C.; Mennel, R.G.; et al. Use of Biomarkers to Guide Decisions on Adjuvant Systemic Therapy for Women With Early-Stage Invasive Breast Cancer: American Society of Clinical Oncology Clinical Practice Guideline. J. Clin. Oncol. 2016, 34, 1134–1150. [Google Scholar] [CrossRef]
  61. Das, S.; Dey, M.K.; Devireddy, R.; Gartia, M.R. Biomarkers in Cancer Detection, Diagnosis, and Prognosis. Sensors 2024, 24, 37. [Google Scholar] [CrossRef]
  62. Cha, Y.; Kim, S.; Han, S.W. Utilizing Plasma Circulating Tumor DNA Sequencing for Precision Medicine in the Management of Solid Cancers. Cancer Res. Treat. 2023, 55, 367–384. [Google Scholar] [CrossRef] [PubMed]
  63. Ko, J.; Bhagwat, N.; Yee, S.S.; Ortiz, N.; Sahmoud, A.; Black, T.; Aiello, N.M.; McKenzie, L.; O’Hara, M.; Redlinger, C.; et al. Combining Machine Learning and Nanofluidic Technology To Diagnose Pancreatic Cancer Using Exosomes. ACS Nano 2017, 11, 11182–11193. [Google Scholar] [CrossRef] [PubMed]
  64. Li, B.; Kugeratski, F.G.; Kalluri, R. A novel machine learning algorithm selects proteome signature to specifically identify cancer exosomes. eLife 2024, 12, RP90390. [Google Scholar] [CrossRef]
  65. Pan, J.; Song, G.; Chen, D.; Li, Y.; Liu, S.; Hu, S.; Rosa, C.; Eichinger, D.; Pino, I.; Zhu, H.; et al. Identification of Serological Biomarkers for Early Diagnosis of Lung Cancer Using a Protein Array-Based Approach. Mol. Cell. Proteom. 2017, 16, 2069–2078. [Google Scholar] [CrossRef] [PubMed]
  66. El-Khoury, V.; Schritz, A.; Kim, S.Y.; Lesur, A.; Sertamo, K.; Bernardin, F.; Petritis, K.; Pirrotte, P.; Selinsky, C.; Whiteaker, J.R.; et al. Identification of a Blood-Based Protein Biomarker Panel for Lung Cancer Detection. Cancers 2020, 12, 1629. [Google Scholar] [CrossRef]
  67. Loomans-Kropp, H.A.; Song, Y.; Gala, M.; Parikh, A.R.; Van Seventer, E.E.; Alvarez, R.; Hitchins, M.P.; Shoemaker, R.H.; Umar, A. Methylated Septin9 (mSEPT9): A promising blood-based biomarker for the detection and screening of early-onset colorectal cancer. Cancer Res. Commun. 2022, 2, 90–98. [Google Scholar] [CrossRef]
  68. Sun, G.; Meng, J.; Duan, H.; Zhang, D.; Tang, Y. Diagnostic Assessment of septin9 DNA Methylation for Colorectal Cancer Using Blood Detection: A Meta-Analysis. Pathol. Oncol. Res. 2019, 25, 1525–1534. [Google Scholar] [CrossRef]
  69. Guo, J.; Liu, D.; Zhang, X.; Johnson, H.; Feng, X.; Zhang, H.; Wu, A.H.B.; Chen, L.; Fang, J.; Xiao, Z.; et al. Establishing a Urine-Based Biomarker Assay for Prostate Cancer Risk Stratification. Front. Cell Dev. Biol. 2020, 8, 597961. [Google Scholar] [CrossRef]
  70. 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]
  71. Curtit, E.; Vannetzel, J.M.; Darmon, J.C.; Roche, S.; Bourgeois, H.; Dewas, S.; Catala, S.; Mereb, E.; Fanget, C.F.; Genet, D.; et al. Results of PONDx, a prospective multicenter study of the Oncotype DX® breast cancer assay: Real-life utilization and decision impact in French clinical practice. Breast 2019, 44, 39–45. [Google Scholar] [CrossRef]
  72. Chen, X.; Gole, J.; Gore, A.; He, Q.; Lu, M.; Min, J.; Yuan, Z.; Yang, X.; Jiang, Y.; Zhang, T.; et al. Non-invasive early detection of cancer four years before conventional diagnosis using a blood test. Nat. Commun. 2020, 11, 3475. [Google Scholar] [CrossRef] [PubMed]
  73. Liu, M.C. Transforming the landscape of early cancer detection using blood tests-Commentary on current methodologies and future prospects. Br. J. Cancer 2021, 124, 1475–1477. [Google Scholar] [CrossRef] [PubMed]
  74. Echle, A.; Rindtorff, N.T.; Brinker, T.J.; Luedde, T.; Pearson, A.T.; Kather, J.N. Deep learning in cancer pathology: A new generation of clinical biomarkers. Br. J. Cancer 2021, 124, 686–696. [Google Scholar] [CrossRef]
  75. Wang, H.Y.; Lin, W.Y.; Zhou, C.; Yang, Z.A.; Kalpana, S.; Lebowitz, M.S. Integrating Artificial Intelligence for Advancing Multiple-Cancer Early Detection via Serum Biomarkers: A Narrative Review. Cancers 2024, 16, 862. [Google Scholar] [CrossRef]
  76. Lee, Y.T.; Tan, Y.J.; Oon, C.E. Molecular targeted therapy: Treating cancer with specificity. Eur. J. Pharmacol. 2018, 834, 188–196. [Google Scholar] [CrossRef] [PubMed]
  77. Wang, M.; Herbst, R.S.; Boshoff, C. Toward personalized treatment approaches for non-small-cell lung cancer. Nat. Med. 2021, 27, 1345–1356. [Google Scholar] [CrossRef]
  78. Gibbs, S.N.; Peneva, D.; Cuyun Carter, G.; Palomares, M.R.; Thakkar, S.; Hall, D.W.; Dalglish, H.; Campos, C.; Yermilov, I. Comprehensive Review on the Clinical Impact of Next-Generation Sequencing Tests for the Management of Advanced Cancer. JCO Precis. Oncol. 2023, 7, e2200715. [Google Scholar] [CrossRef]
  79. Mardis, E.R. The Impact of Next-Generation Sequencing on Cancer Genomics: From Discovery to Clinic. Cold Spring Harb. Perspect. Med. 2019, 9, a036269. [Google Scholar] [CrossRef]
  80. Alharbi, F.; Vakanski, A. Machine Learning Methods for Cancer Classification Using Gene Expression Data: A Review. Bioengineering 2023, 10, 173. [Google Scholar] [CrossRef]
  81. Hayashi, H.; Kurata, T.; Takiguchi, Y.; Arai, M.; Takeda, K.; Akiyoshi, K.; Matsumoto, K.; Onoe, T.; Mukai, H.; Matsubara, N.; et al. Randomized Phase II Trial Comparing Site-Specific Treatment Based on Gene Expression Profiling With Carboplatin and Paclitaxel for Patients With Cancer of Unknown Primary Site. J. Clin. Oncol. 2019, 37, 570–579. [Google Scholar] [CrossRef]
  82. Venetis, K.; Crimini, E.; Sajjadi, E.; Corti, C.; Guerini-Rocco, E.; Viale, G.; Curigliano, G.; Criscitiello, C.; Fusco, N. HER2 Low, Ultra-low, and Novel Complementary Biomarkers: Expanding the Spectrum of HER2 Positivity in Breast Cancer. Front. Mol. Biosci. 2022, 9, 834651. [Google Scholar] [CrossRef] [PubMed]
  83. Karol, S.E.; Yang, J.J. Pharmacogenomics and ALL treatment: How to optimize therapy. Semin. Hematol. 2020, 57, 130–136. [Google Scholar] [CrossRef] [PubMed]
  84. Ding, S.; Liu, J.; Han, X.; Tang, M. CRISPR/Cas9-Mediated Genome Editing in Cancer Therapy. Int. J. Mol. Sci. 2023, 24, 16325. [Google Scholar] [CrossRef]
  85. Li, Y.; Zhou, S.; Wu, Q.; Gong, C. CRISPR/Cas gene editing and delivery systems for cancer therapy. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2024, 16, e1938. [Google Scholar] [CrossRef]
  86. Seebacher, N.A.; Stacy, A.E.; Porter, G.M.; Merlot, A.M. Clinical development of targeted and immune based anti-cancer therapies. J. Exp. Clin. Cancer Res. 2019, 38, 156. [Google Scholar] [CrossRef] [PubMed]
  87. Taghiloo, S.; Norozi, S.; Asgarian-Omran, H. The Effects of PI3K/Akt/mTOR Signaling Pathway Inhibitors on the Expression of Immune Checkpoint Ligands in Acute Myeloid Leukemia Cell Line. Iran. J. Allergy Asthma Immunol. 2022, 21, 178–188. [Google Scholar] [CrossRef]
  88. Setton, J.; Zinda, M.; Riaz, N.; Durocher, D.; Zimmermann, M.; Koehler, M.; Reis-Filho, J.S.; Powell, S.N. Synthetic Lethality in Cancer Therapeutics: The Next Generation. Cancer Discov. 2021, 11, 1626–1635. [Google Scholar] [CrossRef]
  89. Ashworth, A.; Lord, C.J. Synthetic lethal therapies for cancer: What’s next after PARP inhibitors? Nat. Rev. Clin. Oncol. 2018, 15, 564–576. [Google Scholar] [CrossRef]
  90. Hu, Y.; Guo, M. Synthetic lethality strategies: Beyond BRCA1/2 mutations in pancreatic cancer. Cancer Sci. 2020, 111, 3111–3121. [Google Scholar] [CrossRef]
  91. Hosea, R.; Hillary, S.; Wu, S.; Kasim, V. Targeting Transcription Factor YY1 for Cancer Treatment: Current Strategies and Future Directions. Cancers 2023, 15, 3506. [Google Scholar] [CrossRef]
  92. Blass, E.; Ott, P.A. Advances in the development of personalized neoantigen-based therapeutic cancer vaccines. Nat. Rev. Clin. Oncol. 2021, 18, 215–229. [Google Scholar] [CrossRef] [PubMed]
  93. Tan, S.; Li, D.; Zhu, X. Cancer immunotherapy: Pros, cons and beyond. Biomed. Pharmacother. 2020, 124, 109821. [Google Scholar] [CrossRef]
  94. Li, M.; Zhu, J.; Lv, Z.; Qin, H.; Wang, X.; Shi, H. Recent Advances in RNA-Targeted Cancer Therapy. Chembiochem 2024, 25, e202300633. [Google Scholar] [CrossRef] [PubMed]
  95. Haque, S.; Cook, K.; Sahay, G.; Sun, C. RNA-Based Therapeutics: Current Developments in Targeted Molecular Therapy of Triple-Negative Breast Cancer. Pharmaceutics 2021, 13, 1694. [Google Scholar] [CrossRef]
  96. Kang, J.X.; Li, C.; Cheng, Y.M.; Huang, M.X.; Zhao, G.K.; Jin, Z.L.; Qi, X.W.; Gu, J.; Ouyang, Q. Advances in Small-Molecule Dual Inhibitors Targeting EGFR and HER2 Receptors as Anti-Cancer Agents. Curr. Med. Chem. 2024. [Google Scholar] [CrossRef] [PubMed]
  97. Taieb, J.; Jung, A.; Sartore-Bianchi, A.; Peeters, M.; Seligmann, J.; Zaanan, A.; Burdon, P.; Montagut, C.; Laurent-Puig, P. The Evolving Biomarker Landscape for Treatment Selection in Metastatic Colorectal Cancer. Drugs 2019, 79, 1375–1394. [Google Scholar] [CrossRef]
  98. Restrepo, J.C.; Martínez Guevara, D.; Pareja López, A.; Montenegro Palacios, J.F.; Liscano, Y. Identification and Application of Emerging Biomarkers in Treatment of Non-Small-Cell Lung Cancer: Systematic Review. Cancers 2024, 16, 2338. [Google Scholar] [CrossRef]
  99. Hsieh, Y.C.; Kirschner, K.; Copland, M. Improving outcomes in chronic myeloid leukemia through harnessing the immunological landscape. Leukemia 2021, 35, 1229–1242. [Google Scholar] [CrossRef]
  100. O’Shaughnessy, J.; Robert, N.; Annavarapu, S.; Zhou, J.; Sussell, J.; Cheng, A.; Fung, A. Recurrence rates in patients with HER2+ breast cancer who achieved a pathological complete response after neoadjuvant pertuzumab plus trastuzumab followed by adjuvant trastuzumab: A real-world evidence study. Breast Cancer Res. Treat. 2021, 187, 903–913. [Google Scholar] [CrossRef]
  101. Huo, K.G.; Notsuda, H.; Fang, Z.; Liu, N.F.; Gebregiworgis, T.; Li, Q.; Pham, N.A.; Li, M.; Liu, N.; Shepherd, F.A.; et al. Lung Cancer Driven by BRAF(G469V) Mutation Is Targetable by EGFR Kinase Inhibitors. J. Thorac. Oncol. 2022, 17, 277–288. [Google Scholar] [CrossRef]
  102. Planchard, D.; Jänne, P.A.; Cheng, Y.; Yang, J.C.; Yanagitani, N.; Kim, S.W.; Sugawara, S.; Yu, Y.; Fan, Y.; Geater, S.L.; et al. Osimertinib with or without Chemotherapy in EGFR-Mutated Advanced NSCLC. N. Engl. J. Med. 2023, 389, 1935–1948. [Google Scholar] [CrossRef] [PubMed]
  103. Gambardella, V.; Tarazona, N.; Cejalvo, J.M.; Lombardi, P.; Huerta, M.; Roselló, S.; Fleitas, T.; Roda, D.; Cervantes, A. Personalized Medicine: Recent Progress in Cancer Therapy. Cancers 2020, 12, 1009. [Google Scholar] [CrossRef] [PubMed]
  104. Walcher, L.; Kistenmacher, A.K.; Suo, H.; Kitte, R.; Dluczek, S.; Strauß, A.; Blaudszun, A.R.; Yevsa, T.; Fricke, S.; Kossatz-Boehlert, U. Cancer Stem Cells-Origins and Biomarkers: Perspectives for Targeted Personalized Therapies. Front. Immunol. 2020, 11, 1280. [Google Scholar] [CrossRef] [PubMed]
  105. Mitra, A.K.; Agrahari, V.; Mandal, A.; Cholkar, K.; Natarajan, C.; Shah, S.; Joseph, M.; Trinh, H.M.; Vaishya, R.; Yang, X.; et al. Novel delivery approaches for cancer therapeutics. J. Control Release 2015, 219, 248–268. [Google Scholar] [CrossRef]
  106. Bankó, P.; Lee, S.Y.; Nagygyörgy, V.; Zrínyi, M.; Chae, C.H.; Cho, D.H.; Telekes, A. Technologies for circulating tumor cell separation from whole blood. J. Hematol. Oncol. 2019, 12, 48. [Google Scholar] [CrossRef]
  107. O’Connor, O.; McVeigh, T.P. Increasing use of artificial intelligence in genomic medicine for cancer care- the promise and potential pitfalls. BJC Rep. 2025, 3, 20. [Google Scholar] [CrossRef]
  108. Rodriguez, H.; Zenklusen, J.C.; Staudt, L.M.; Doroshow, J.H.; Lowy, D.R. The next horizon in precision oncology: Proteogenomics to inform cancer diagnosis and treatment. Cell 2021, 184, 1661–1670. [Google Scholar] [CrossRef]
  109. Boehm, K.M.; Khosravi, P.; Vanguri, R.; Gao, J.; Shah, S.P. Harnessing multimodal data integration to advance precision oncology. Nat. Rev. Cancer 2022, 22, 114–126. [Google Scholar] [CrossRef]
  110. 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]
  111. Zubair, M.; Wang, S.; Ali, N. Advanced Approaches to Breast Cancer Classification and Diagnosis. Front. Pharmacol. 2020, 11, 632079. [Google Scholar] [CrossRef]
  112. Dienstmann, R.; Jang, I.S.; Bot, B.; Friend, S.; Guinney, J. Database of genomic biomarkers for cancer drugs and clinical targetability in solid tumors. Cancer Discov. 2015, 5, 118–123. [Google Scholar] [CrossRef] [PubMed]
  113. Kamel, H.F.M.; Al-Amodi, H. Exploitation of Gene Expression and Cancer Biomarkers in Paving the Path to Era of Personalized Medicine. Genom. Proteom. Bioinform. 2017, 15, 220–235. [Google Scholar] [CrossRef] [PubMed]
  114. Song, Z.; Lian, S.; Mak, S.; Chow, M.Z.; Xu, C.; Wang, W.; Keung, H.Y.; Lu, C.; Kebede, F.T.; Gao, Y.; et al. Deep RNA Sequencing Revealed Fusion Junctional Heterogeneity May Predict Crizotinib Treatment Efficacy in ALK-Rearranged NSCLC. J. Thorac. Oncol. 2022, 17, 264–276. [Google Scholar] [CrossRef]
  115. Yaeger, R.; Weiss, J.; Pelster, M.S.; Spira, A.I.; Barve, M.; Ou, S.I.; Leal, T.A.; Bekaii-Saab, T.S.; Paweletz, C.P.; Heavey, G.A.; et al. Adagrasib with or without Cetuximab in Colorectal Cancer with Mutated KRAS G12C. N. Engl. J. Med. 2023, 388, 44–54. [Google Scholar] [CrossRef] [PubMed]
  116. Chen, P.; Li, X.; Zhang, R.; Liu, S.; Xiang, Y.; Zhang, M.; Chen, X.; Pan, T.; Yan, L.; Feng, J.; et al. Combinative treatment of β-elemene and cetuximab is sensitive to KRAS mutant colorectal cancer cells by inducing ferroptosis and inhibiting epithelial-mesenchymal transformation. Theranostics 2020, 10, 5107–5119. [Google Scholar] [CrossRef]
  117. Fernandez, G.; Prastawa, M.; Madduri, A.S.; Scott, R.; Marami, B.; Shpalensky, N.; Cascetta, K.; Sawyer, M.; Chan, M.; Koll, G.; et al. Development and validation of an AI-enabled digital breast cancer assay to predict early-stage breast cancer recurrence within 6 years. Breast Cancer Res. 2022, 24, 93. [Google Scholar] [CrossRef]
  118. Jeong, S.Y.; Chung, J.Y.; Byeon, S.J.; Kim, C.J.; Lee, Y.Y.; Kim, T.J.; Lee, J.W.; Kim, B.G.; Chae, Y.L.; Oh, S.Y.; et al. Validation of Potential Protein Markers Predicting Chemoradioresistance in Early Cervical Cancer by Immunohistochemistry. Front. Oncol. 2021, 11, 665595. [Google Scholar] [CrossRef]
  119. Khattak, M.A.; Reid, A.; Freeman, J.; Pereira, M.; McEvoy, A.; Lo, J.; Frank, M.H.; Meniawy, T.; Didan, A.; Spencer, I.; et al. PD-L1 Expression on Circulating Tumor Cells May Be Predictive of Response to Pembrolizumab in Advanced Melanoma: Results from a Pilot Study. Oncologist 2020, 25, e520–e527. [Google Scholar] [CrossRef]
  120. Incorvaia, L.; Rinaldi, G.; Badalamenti, G.; Cucinella, A.; Brando, C.; Madonia, G.; Fiorino, A.; Pipitone, A.; Perez, A.; Li Pomi, F.; et al. Prognostic role of soluble PD-1 and BTN2A1 in overweight melanoma patients treated with nivolumab or pembrolizumab: Finding the missing links in the symbiotic immune-metabolic interplay. Ther. Adv. Med. Oncol. 2023, 15, 17588359231151845. [Google Scholar] [CrossRef]
  121. Camidge, D.R.; Doebele, R.C.; Kerr, K.M. Comparing and contrasting predictive biomarkers for immunotherapy and targeted therapy of NSCLC. Nat. Rev. Clin. Oncol. 2019, 16, 341–355. [Google Scholar] [CrossRef]
  122. Jackson, S.E.; Chester, J.D. Personalised cancer medicine. Int. J. Cancer 2015, 137, 262–266. [Google Scholar] [CrossRef] [PubMed]
  123. Nnaji, C.A.; Kuodi, P.; Walter, F.M.; Moodley, J. Effectiveness of interventions for improving timely diagnosis of breast and cervical cancers in low-income and middle-income countries: A systematic review. BMJ Open 2022, 12, e054501. [Google Scholar] [CrossRef] [PubMed]
  124. Fulton, J.J.; LeBlanc, T.W.; Cutson, T.M.; Porter Starr, K.N.; Kamal, A.; Ramos, K.; Freiermuth, C.E.; McDuffie, J.R.; Kosinski, A.; Adam, S.; et al. Integrated outpatient palliative care for patients with advanced cancer: A systematic review and meta-analysis. Palliat. Med. 2019, 33, 123–134. [Google Scholar] [CrossRef] [PubMed]
  125. Balitsky, A.K.; Rayner, D.; Britto, J.; Lionel, A.C.; Ginsberg, L.; Cho, W.; Wilfred, A.M.; Sardar, H.; Cantor, N.; Mian, H.; et al. Patient-Reported Outcome Measures in Cancer Care: An Updated Systematic Review and Meta-Analysis. JAMA Netw. Open 2024, 7, e2424793. [Google Scholar] [CrossRef]
  126. Liu, Q.; Wang, C.; Zheng, Y.; Zhao, Y.; Wang, Y.; Hao, J.; Zhao, X.; Yi, K.; Shi, L.; Kang, C.; et al. Virus-like nanoparticle as a co-delivery system to enhance efficacy of CRISPR/Cas9-based cancer immunotherapy. Biomaterials 2020, 258, 120275. [Google Scholar] [CrossRef]
  127. Aghamiri, S.; Talaei, S.; Ghavidel, A.A.; Zandsalimi, F.; Masoumi, S.; Hafshejani, N.H.; Jajarmi, V. Nanoparticles-mediated CRISPR/Cas9 delivery: Recent advances in cancer treatment. J. Drug Deliv. Sci. Technol. 2020, 56, 101533. [Google Scholar] [CrossRef]
  128. Mao, X.; Wu, S.; Huang, D.; Li, C. Complications and comorbidities associated with antineoplastic chemotherapy: Rethinking drug design and delivery for anticancer therapy. Acta Pharm. Sin. B 2024, 14, 2901–2926. [Google Scholar] [CrossRef]
  129. Hou, J.J.; Maithel, S.K.; Weber, S.M.; Poultsides, G.; Wolfgang, C.L.; Fields, R.C.; He, J.; Scoggins, C.; Idrees, K.; Shen, P.; et al. Impact of adjuvant therapy on outcomes after curative-intent resection for distal cholangiocarcinoma. J. Surg. Oncol. 2023, 127, 607–615. [Google Scholar] [CrossRef]
  130. Olson, D.J.; Eroglu, Z.; Brockstein, B.; Poklepovic, A.S.; Bajaj, M.; Babu, S.; Hallmeyer, S.; Velasco, M.; Lutzky, J.; Higgs, E.; et al. Pembrolizumab Plus Ipilimumab Following Anti-PD-1/L1 Failure in Melanoma. J. Clin. Oncol. 2021, 39, 2647–2655. [Google Scholar] [CrossRef]
  131. Yu, H.A.; Schoenfeld, A.J.; Makhnin, A.; Kim, R.; Rizvi, H.; Tsui, D.; Falcon, C.; Houck-Loomis, B.; Meng, F.; Yang, J.L.; et al. Effect of Osimertinib and Bevacizumab on Progression-Free Survival for Patients with Metastatic EGFR-Mutant Lung Cancers: A Phase 1/2 Single-Group Open-Label Trial. JAMA Oncol. 2020, 6, 1048–1054. [Google Scholar] [CrossRef]
  132. O’Shaughnessy, J.; Graham, C.L.; Whitworth, P.W.; Beitsch, P.; Osborne, C.R.C.; Layeequr Rahman, R.; Brown, E.A.; Gold, L.P.; Johnson, N.M.; Brufsky, A.M.; et al. Association of MammaPrint index and 3-year outcome of patients with HR+HER2- early-stage breast cancer treated with chemotherapy with or without anthracycline. J. Clin. Oncol. 2024, 42, 511. [Google Scholar] [CrossRef]
  133. Guarneri, V.; Griguolo, G.; Miglietta, F.; Conte, P.F.; Dieci, M.V.; Girardi, F. Survival after neoadjuvant therapy with trastuzumab-lapatinib and chemotherapy in patients with HER2-positive early breast cancer: A meta-analysis of randomized trials. ESMO Open 2022, 7, 100433. [Google Scholar] [CrossRef] [PubMed]
  134. Solomon, B.J.; Loong, H.H.; Summers, Y.; Thomas, Z.M.; French, P.; Lin, B.K.; Sashegyi, A.; Wolf, J.; Yang, J.C.; Drilon, A. Correlation between treatment effects on response rate and progression-free survival and overall survival in trials of targeted therapies in molecularly enriched populations. ESMO Open 2022, 7, 100398. [Google Scholar] [CrossRef] [PubMed]
  135. Tsimberidou, A.M.; Hong, D.S.; Fu, S.; Karp, D.D.; Piha-Paul, S.; Kies, M.S.; Ravi, V.; Subbiah, V.; Patel, S.M.; Tu, S.M.; et al. Precision medicine: Preliminary results from the Initiative for Molecular Profiling and Advanced Cancer Therapy 2 (IMPACT2) study. NPJ Precis. Oncol. 2021, 5, 21. [Google Scholar] [CrossRef]
  136. Neagu, A.N.; Bruno, P.; Johnson, K.R.; Ballestas, G.; Darie, C.C. Biological Basis of Breast Cancer-Related Disparities in Precision Oncology Era. Int. J. Mol. Sci. 2024, 25, 4113. [Google Scholar] [CrossRef]
  137. Aldea, M.; Andre, F.; Marabelle, A.; Dogan, S.; Barlesi, F.; Soria, J.C. Overcoming Resistance to Tumor-Targeted and Immune-Targeted Therapies. Cancer Discov. 2021, 11, 874–899. [Google Scholar] [CrossRef]
  138. Sharma, K.; Mayer, T.; Li, S.; Qureshi, S.; Farooq, F.; Vuylsteke, P.; Ralefala, T.; Marlink, R. Advancing oncology drug therapies for sub-Saharan Africa. PLOS Glob. Public Health 2023, 3, e0001653. [Google Scholar] [CrossRef] [PubMed]
  139. Twahir, M.; Oyesegun, R.; Yarney, J.; Gachii, A.; Edusa, C.; Nwogu, C.; Mangutha, G.; Anderson, P.; Benjamin, E.; Müller, B.; et al. Real-world challenges for patients with breast cancer in sub-Saharan Africa: A retrospective observational study of access to care in Ghana, Kenya and Nigeria. BMJ Open 2021, 11, e041900. [Google Scholar] [CrossRef]
  140. Fu, Y.; Saraswat, A.; Wei, Z.; Agrawal, M.Y.; Dukhande, V.V.; Reznik, S.E.; Patel, K. Development of Dual ARV-825 and Nintedanib-Loaded PEGylated Nano-Liposomes for Synergistic Efficacy in Vemurafnib-Resistant Melanoma. Pharmaceutics 2021, 13, 1005. [Google Scholar] [CrossRef]
  141. Wang, S.; Tsui, S.T.; Liu, C.; Song, Y.; Liu, D. EGFR C797S mutation mediates resistance to third-generation inhibitors in T790M-positive non-small cell lung cancer. J. Hematol. Oncol. 2016, 9, 59. [Google Scholar] [CrossRef]
  142. Yu, H.A.; Tian, S.K.; Drilon, A.E.; Borsu, L.; Riely, G.J.; Arcila, M.E.; Ladanyi, M. Acquired Resistance of EGFR-Mutant Lung Cancer to a T790M-Specific EGFR Inhibitor: Emergence of a Third Mutation (C797S) in the EGFR Tyrosine Kinase Domain. JAMA Oncol. 2015, 1, 982–984. [Google Scholar] [CrossRef] [PubMed]
  143. Liu, D. Cancer biomarkers for targeted therapy. Biomark. Res. 2019, 7, 25. [Google Scholar] [CrossRef]
  144. Dong, P.; Mao, A.; Qiu, W.; Li, G. Improvement of Cancer Prevention and Control: Reflection on the Role of Emerging Information Technologies. J. Med. Internet Res. 2024, 26, e50000. [Google Scholar] [CrossRef] [PubMed]
  145. Hesse, B.W.; Kwasnicka, D.; Ahern, D.K. Emerging digital technologies in cancer treatment, prevention, and control. Transl. Behav. Med. 2021, 11, 2009–2017. [Google Scholar] [CrossRef]
  146. Fu, S.W.; Tang, C.; Tan, X.; Srivastava, S. Liquid biopsy for early cancer detection: Technological revolutions and clinical dilemma. Expert. Rev. Mol. Diagn. 2024, 24, 937–955. [Google Scholar] [CrossRef]
  147. Yu, W.; Hurley, J.; Roberts, D.; Chakrabortty, S.K.; Enderle, D.; Noerholm, M.; Breakefield, X.O.; Skog, J.K. Exosome-based liquid biopsies in cancer: Opportunities and challenges. Ann. Oncol. 2021, 32, 466–477. [Google Scholar] [CrossRef] [PubMed]
  148. Jin, D.; Khan, N.U.; Gu, W.; Lei, H.; Goel, A.; Chen, T. Informatics strategies for early detection and risk mitigation in pancreatic cancer patients. Neoplasia 2025, 60, 101129. [Google Scholar] [CrossRef]
  149. Rolfo, C.; Russo, A. Moving Forward Liquid Biopsy in Early Liver Cancer Detection. Cancer Discov. 2023, 13, 532–534. [Google Scholar] [CrossRef]
  150. Milner, D.A., Jr.; Lennerz, J.K. Technology and Future of Multi-Cancer Early Detection. Life 2024, 14, 833. [Google Scholar] [CrossRef]
  151. Passaro, A.; Al Bakir, M.; Hamilton, E.G.; Diehn, M.; André, 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]
  152. Lim, Z.F.; Ma, P.C. Emerging insights of tumor heterogeneity and drug resistance mechanisms in lung cancer targeted therapy. J. Hematol. Oncol. 2019, 12, 134. [Google Scholar] [CrossRef] [PubMed]
  153. Recondo, G.; Che, J.; Jänne, P.A.; Awad, M.M. Targeting MET Dysregulation in Cancer. Cancer Discov. 2020, 10, 922–934. [Google Scholar] [CrossRef] [PubMed]
  154. Chen, M.; Zhao, H. Next-generation sequencing in liquid biopsy: Cancer screening and early detection. Hum. Genom. 2019, 13, 34. [Google Scholar] [CrossRef] [PubMed]
  155. El-Tanani, M.; Rabbani, S.A.; Babiker, R.; Rangraze, I.; Kapre, S.; Palakurthi, S.S.; Alnuqaydan, A.M.; Aljabali, A.A.; Rizzo, M.; El-Tanani, Y.; et al. Unraveling the tumor microenvironment: Insights into cancer metastasis and therapeutic strategies. Cancer Lett. 2024, 591, 216894. [Google Scholar] [CrossRef]
  156. Che, G.; Yin, J.; Wang, W.; Luo, Y.; Chen, Y.; Yu, X.; Wang, H.; Liu, X.; Chen, Z.; Wang, X.; et al. Circumventing drug resistance in gastric cancer: A spatial multi-omics exploration of chemo and immuno-therapeutic response dynamics. Drug Resist. Updat. 2024, 74, 101080. [Google Scholar] [CrossRef]
  157. Lei, Z.N.; Tian, Q.; Teng, Q.X.; Wurpel, J.N.D.; Zeng, L.; Pan, Y.; Chen, Z.S. Understanding and targeting resistance mechanisms in cancer. MedComm 2023, 4, e265. [Google Scholar] [CrossRef]
Figure 1. Illustration of a cancer cell surrounded by various biomarkers (DNA, RNA, mRNAs, proteins and enzymes), utilized in cancer diagnosis. Each biomarker plays a crucial role in identifying specific cancer types and guiding personalized treatment strategies. The picture was assembled using dynamic BioRender (https://www.biorender.com/ Accessed on 28 March 2025).
Figure 1. Illustration of a cancer cell surrounded by various biomarkers (DNA, RNA, mRNAs, proteins and enzymes), utilized in cancer diagnosis. Each biomarker plays a crucial role in identifying specific cancer types and guiding personalized treatment strategies. The picture was assembled using dynamic BioRender (https://www.biorender.com/ Accessed on 28 March 2025).
Jmp 06 00020 g001
Figure 2. Comparison of personalized molecular treatments and AI-driven therapies versus conventional treatment techniques in cancer care. This illustration highlights the advancements in precision medicine that aim to improve efficacy and reduce side effects for patients. The picture was assembled using dynamic BioRender.
Figure 2. Comparison of personalized molecular treatments and AI-driven therapies versus conventional treatment techniques in cancer care. This illustration highlights the advancements in precision medicine that aim to improve efficacy and reduce side effects for patients. The picture was assembled using dynamic BioRender.
Jmp 06 00020 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

Molla, G.; Bitew, M. The Future of Cancer Diagnosis and Treatment: Unlocking the Power of Biomarkers and Personalized Molecular-Targeted Therapies. J. Mol. Pathol. 2025, 6, 20. https://doi.org/10.3390/jmp6030020

AMA Style

Molla G, Bitew M. The Future of Cancer Diagnosis and Treatment: Unlocking the Power of Biomarkers and Personalized Molecular-Targeted Therapies. Journal of Molecular Pathology. 2025; 6(3):20. https://doi.org/10.3390/jmp6030020

Chicago/Turabian Style

Molla, Getnet, and Molalegne Bitew. 2025. "The Future of Cancer Diagnosis and Treatment: Unlocking the Power of Biomarkers and Personalized Molecular-Targeted Therapies" Journal of Molecular Pathology 6, no. 3: 20. https://doi.org/10.3390/jmp6030020

APA Style

Molla, G., & Bitew, M. (2025). The Future of Cancer Diagnosis and Treatment: Unlocking the Power of Biomarkers and Personalized Molecular-Targeted Therapies. Journal of Molecular Pathology, 6(3), 20. https://doi.org/10.3390/jmp6030020

Article Metrics

Back to TopTop