Appearance
This report is written by MaltSci based on the latest literature and research findings
How does targeted therapy work in precision oncology?
Abstract
Cancer remains a leading cause of morbidity and mortality globally, necessitating advancements in therapeutic strategies. Traditional chemotherapy approaches have significant limitations, prompting a shift towards precision oncology, which tailors treatment based on individual tumor characteristics. Targeted therapy is a cornerstone of this paradigm, employing agents that inhibit key molecular pathways involved in tumor growth, thus improving efficacy while minimizing damage to normal cells. This report provides an overview of targeted therapy mechanisms, including pathway inhibition, antibody-drug conjugates, immune checkpoint inhibitors, and gene expression modulation. It also emphasizes the critical role of biomarkers in patient selection, enhancing treatment personalization. Clinical applications across breast cancer, lung cancer, and melanoma illustrate the benefits of targeted therapies, although challenges such as tumor heterogeneity and resistance mechanisms persist. Moreover, socio-economic barriers can hinder access to these innovative treatments. Looking ahead, the field is poised for growth through novel target identification, combination therapies, and personalized medicine approaches. Continued research is essential to address current limitations and improve patient outcomes in cancer treatment.
Outline
This report will discuss the following questions.
- 1 Introduction
- 2 Mechanisms of Targeted Therapy
- 2.1 Overview of Targeted Therapy Mechanisms
- 2.2 Role of Biomarkers in Targeted Therapy
- 3 Clinical Applications of Targeted Therapy
- 3.1 Targeted Therapies in Breast Cancer
- 3.2 Targeted Therapies in Lung Cancer
- 3.3 Targeted Therapies in Melanoma
- 4 Challenges and Limitations
- 4.1 Tumor Heterogeneity
- 4.2 Resistance Mechanisms
- 4.3 Access and Affordability
- 5 Future Directions in Targeted Therapy
- 5.1 Novel Target Identification
- 5.2 Combination Therapies
- 5.3 Personalized Medicine Approaches
- 6 Summary
1 Introduction
Cancer remains one of the leading causes of morbidity and mortality worldwide, necessitating continuous advancements in therapeutic strategies. The traditional approach to cancer treatment, primarily based on cytotoxic chemotherapy, has faced significant limitations, including non-specificity, systemic toxicity, and the emergence of drug resistance. In recent years, the paradigm has shifted towards precision oncology, a transformative approach that tailors treatment based on the individual genetic and molecular characteristics of a patient's tumor. Targeted therapy, a cornerstone of precision oncology, employs specific agents to inhibit key molecular pathways involved in tumor growth and survival, thereby improving therapeutic efficacy while minimizing damage to normal cells [1].
The significance of targeted therapy in oncology cannot be overstated. By focusing on the unique genetic alterations present in a patient's tumor, targeted therapies aim to optimize treatment outcomes and reduce adverse effects associated with conventional therapies. This personalized approach not only enhances the therapeutic index but also facilitates the identification of biomarkers that can guide clinical decision-making [2]. Moreover, as our understanding of cancer biology deepens, the potential for targeted therapies to address the heterogeneity of tumors becomes increasingly apparent, allowing for more effective management of various malignancies, including breast cancer, lung cancer, and melanoma [3].
Currently, the landscape of targeted therapy is evolving rapidly, with numerous agents receiving regulatory approval and entering clinical practice. The advent of next-generation sequencing technologies has enabled comprehensive molecular profiling of tumors, leading to the identification of actionable mutations and guiding the selection of appropriate therapies [4]. However, despite the promising advances, challenges remain. Tumor heterogeneity, resistance mechanisms, and issues related to access and affordability continue to hinder the widespread implementation of targeted therapies [5]. Furthermore, while some therapies have demonstrated significant efficacy, others have shown limited benefits, highlighting the need for continued research and innovation in this field [6].
This report aims to provide a comprehensive overview of how targeted therapy operates within the framework of precision oncology. The content is organized as follows: first, we will explore the mechanisms of targeted therapy, including an overview of the various strategies employed and the critical role of biomarkers in patient selection. Next, we will examine the clinical applications of targeted therapies across different cancer types, specifically focusing on breast cancer, lung cancer, and melanoma. Following this, we will discuss the challenges and limitations associated with targeted therapies, including tumor heterogeneity, resistance mechanisms, and the socio-economic barriers that may impede access to these treatments. Finally, we will look towards the future of targeted therapy, considering novel target identification, the potential of combination therapies, and the promise of personalized medicine approaches.
By elucidating the mechanisms, applications, and challenges of targeted therapies, this report seeks to contribute to the ongoing discourse in precision oncology and underscore the importance of continued research in improving patient outcomes in cancer treatment.
2 Mechanisms of Targeted Therapy
2.1 Overview of Targeted Therapy Mechanisms
Targeted therapy in precision oncology is a sophisticated approach that focuses on the molecular and genetic characteristics of tumors, allowing for a more personalized treatment strategy. This methodology is designed to specifically inhibit key molecular mechanisms or pathways that are responsible for cancer growth and progression, thereby enhancing treatment efficacy while minimizing damage to normal cells.
The mechanisms of targeted therapy can be broadly categorized into several types:
Inhibition of Specific Pathways: Targeted therapies often involve the use of small molecules or monoclonal antibodies that interfere with specific signaling pathways that are dysregulated in cancer. For instance, these therapies can block growth factor receptors or downstream signaling molecules that are critical for tumor cell proliferation and survival. By inhibiting these pathways, targeted therapies can effectively halt tumor growth and induce apoptosis in cancer cells. This approach has been exemplified in therapies targeting the epidermal growth factor receptor (EGFR) and the human epidermal growth factor receptor 2 (HER2) in breast cancer [3].
Antibody-Drug Conjugates (ADCs): These are a class of targeted therapies that combine the specificity of monoclonal antibodies with the cytotoxicity of chemotherapeutic agents. ADCs deliver cytotoxic drugs directly to cancer cells that express specific antigens, thereby enhancing the therapeutic index and reducing systemic toxicity. The development of ADCs has been a significant advancement in targeted cancer therapy [7].
Gene Expression Modulation: Another mechanism involves the use of antibody-oligonucleotide conjugates (AOCs), which are designed to regulate gene expression. AOCs combine antibodies with oligonucleotides to selectively target and modulate disease-causing proteins, thereby addressing the genetic alterations that drive tumorigenesis [8].
Immune Checkpoint Inhibition: Targeted therapies also include immune checkpoint inhibitors that enhance the body’s immune response against cancer cells. By blocking proteins that inhibit T-cell activation, these therapies can reinvigorate the immune system to recognize and destroy cancer cells. This has been a transformative approach in the treatment of various malignancies [7].
Exogenous and Endogenous Stimuli-Responsive Prodrugs: Precision chemotherapy can also utilize stimuli-responsive prodrugs that are activated in the tumor microenvironment. These prodrugs can be triggered by external factors (like light or ultrasound) or internal factors (such as hypoxia or specific enzyme overexpression), allowing for localized activation of the drug and minimizing off-target effects [9].
Mitochondria-Targeted Therapies: Recent advancements have highlighted the potential of mitochondria-targeted therapies, which focus on delivering therapeutic agents directly to mitochondria. This approach can disrupt mitochondrial functions critical for cancer cell survival, thereby enhancing the therapeutic effect while reducing systemic toxicity [10].
Functional Precision Medicine: This innovative strategy involves testing live tumor cells with various drugs to identify specific vulnerabilities, thus guiding therapy based on real-time responses rather than static genetic profiles. This method acknowledges the heterogeneity of tumors and aims to provide personalized treatment options [11].
In summary, targeted therapy in precision oncology leverages a deep understanding of the molecular underpinnings of cancer to devise treatment strategies that are not only more effective but also tailored to the individual characteristics of a patient’s tumor. This approach is continually evolving, with ongoing research aimed at overcoming current limitations and enhancing the therapeutic landscape for cancer patients [3][12][13].
2.2 Role of Biomarkers in Targeted Therapy
Targeted therapy in precision oncology operates on the principle of customizing treatment based on the unique genetic and molecular characteristics of an individual’s tumor. This approach aims to enhance therapeutic efficacy while minimizing adverse effects, ultimately improving patient outcomes. The mechanisms underlying targeted therapy involve the identification and inhibition of specific molecular targets associated with cancer cell proliferation and survival.
The cornerstone of targeted therapy is the utilization of biomarkers, which are biological indicators that can provide insights into the tumor's characteristics. Biomarkers play a crucial role in the selection of appropriate therapies, as they can indicate which patients are likely to benefit from specific treatments. For instance, in colorectal cancer (CRC), biomarkers such as microsatellite instability (MSI), RAS, and BRAF mutations guide treatment decisions. Emerging biomarkers, including HER-2, consensus molecular subtypes (CMS), and circulating tumor DNA (ctDNA), are being explored for their potential to enhance precision medicine in CRC [14].
The identification of predictive biomarkers enables the stratification of patients into subgroups that are more likely to respond to particular therapies. This stratification is essential for optimizing treatment regimens and improving response rates. For example, targeted therapies such as anti-EGFR and anti-VEGF are employed based on the presence of specific genetic alterations [15]. Furthermore, the development of companion diagnostics, which are tests designed to identify biomarkers that predict response to a particular therapy, is integral to the success of targeted treatments [2].
The mechanism of action of targeted therapies typically involves the inhibition of key signaling pathways that are crucial for tumor growth and survival. For instance, therapies targeting the epidermal growth factor receptor (EGFR) can block signals that promote cell division and tumor growth. Additionally, immune checkpoint inhibitors can enhance the body’s immune response against cancer cells by blocking proteins that suppress immune activity [16].
As precision oncology continues to evolve, the integration of advanced molecular profiling techniques, such as next-generation sequencing, has significantly enhanced the ability to identify actionable mutations and corresponding targeted therapies [17]. However, challenges remain, including the need for comprehensive biomarker validation, the management of drug resistance, and the ethical considerations surrounding the costs and accessibility of precision therapies [18].
In conclusion, targeted therapy in precision oncology is a sophisticated approach that relies heavily on biomarkers to guide treatment decisions. By focusing on the molecular characteristics of tumors, targeted therapies aim to provide more effective and personalized treatment options, ultimately leading to better patient outcomes in the fight against cancer.
3 Clinical Applications of Targeted Therapy
3.1 Targeted Therapies in Breast Cancer
Targeted therapy in precision oncology represents a significant advancement in the treatment of breast cancer by focusing on specific molecular and genetic alterations within tumors. This approach is rooted in the understanding that cancer is a highly heterogeneous disease, and that each patient's tumor possesses unique characteristics that can be exploited for therapeutic benefit.
The principle of targeted therapy is to inhibit key molecular mechanisms or pathways that are crucial for cancer cell survival and proliferation. For breast cancer, various targeted therapies have been developed, each designed to interfere with specific targets. For instance, the use of drugs that target the HER2 protein, such as trastuzumab (Herceptin), exemplifies how targeted therapies can be employed to improve outcomes for patients with HER2-positive breast cancer. Trastuzumab works by binding to the HER2 receptor on the surface of cancer cells, blocking their growth signals and marking them for destruction by the immune system [1].
Moreover, precision oncology aims to tailor treatment based on the individual patient's genetic profile. The identification of predictive biomarkers plays a crucial role in this process. Predictive biomarkers can guide clinical decision-making, enabling oncologists to select therapies that are most likely to be effective based on the genetic alterations present in a patient's tumor. This has led to the development of companion diagnostics that accompany targeted therapies, allowing for more personalized treatment regimens [2].
However, challenges remain in the implementation of targeted therapies in breast cancer. While some therapies have shown remarkable efficacy, many others lack clear biomarkers, which limits their applicability. This disparity creates a division between the "haves" and "have nots" in precision oncology, where only certain therapies can be effectively matched to patient-specific tumor characteristics [2]. Furthermore, the complexity of breast cancer often results in treatment resistance, necessitating ongoing research into alternative strategies, such as drug repurposing and combination therapies, to overcome these hurdles [5].
In conclusion, targeted therapy in precision oncology for breast cancer operates through the identification and inhibition of specific molecular targets associated with tumor growth and survival. The effectiveness of these therapies is significantly enhanced by the integration of predictive biomarkers, which facilitate personalized treatment approaches. Despite the promise of targeted therapies, ongoing challenges, including the need for robust biomarkers and the emergence of resistance, highlight the importance of continued research and innovation in this field.
3.2 Targeted Therapies in Lung Cancer
Targeted therapy in precision oncology operates on the principle of customizing cancer treatment based on the unique molecular and genetic characteristics of an individual’s tumor. This approach is predicated on the understanding that cancer is not a uniform disease; rather, it is highly heterogeneous, with each tumor exhibiting distinct genetic variations that can influence its behavior and response to treatment.
In the context of lung cancer, targeted therapies specifically aim to inhibit key molecular pathways or genetic alterations that drive tumor growth and survival. For instance, the identification of specific mutations, such as those in the epidermal growth factor receptor (EGFR) or anaplastic lymphoma kinase (ALK), has led to the development of targeted agents that can effectively block these aberrant signals. The goal is to improve treatment efficacy while minimizing adverse effects typically associated with conventional chemotherapy.
Several targeted therapies have been approved for lung cancer, particularly for non-small cell lung cancer (NSCLC). For example, EGFR inhibitors like erlotinib and gefitinib are utilized for patients whose tumors harbor EGFR mutations. Similarly, ALK inhibitors, such as crizotinib, are effective in patients with ALK rearrangements. These therapies are guided by companion diagnostics, which are tests that identify the presence of specific biomarkers in the tumor, thus determining eligibility for these targeted treatments.
However, despite the advancements in targeted therapy, challenges remain. A significant proportion of lung cancer patients may not have identifiable mutations that can be targeted, leading to what is referred to as the "have nots" of precision oncology. This underscores the necessity for ongoing research to identify new biomarkers and develop additional targeted therapies that can benefit a broader range of patients. Moreover, the emergence of resistance to targeted therapies poses a significant challenge, as tumors may adapt and develop new mutations that render existing treatments ineffective.
In summary, targeted therapy in precision oncology for lung cancer is a dynamic and evolving field. It focuses on the precise identification of molecular targets within tumors, allowing for the development of tailored therapeutic strategies that enhance treatment efficacy and improve patient outcomes. Continuous research efforts are essential to expand the repertoire of available targeted therapies and address the complexities associated with tumor heterogeneity and resistance mechanisms[2][12][19].
3.3 Targeted Therapies in Melanoma
Targeted therapy in precision oncology operates by specifically inhibiting the growth of cancer cells through the exploitation of unique molecular and genetic characteristics of tumors. In the context of melanoma, targeted therapies have made significant strides in improving patient outcomes, particularly through the identification of specific genetic mutations and alterations that drive the disease.
One of the primary targets in melanoma is the BRAF gene, which, when mutated, is found in approximately 40-60% of melanoma cases. Targeted therapies, such as BRAF inhibitors (e.g., vemurafenib) and MEK inhibitors (e.g., trametinib), have been developed to selectively inhibit the activity of the BRAF protein, leading to reduced tumor growth and improved survival rates for patients with BRAF-mutated melanoma [20]. The successful implementation of these therapies has transformed the treatment landscape for melanoma, significantly extending median survival from around 9 months to over 25 months in some cases [21].
In addition to BRAF-targeted therapies, precision oncology incorporates a variety of other strategies that focus on the molecular profile of individual tumors. For instance, therapies targeting less common genetic alterations, such as NTRK fusions or KIT mutations, are increasingly being utilized to treat specific melanoma subtypes [22]. This approach emphasizes the importance of comprehensive genomic profiling, which allows clinicians to tailor treatments based on the unique genetic makeup of a patient's tumor.
Moreover, the integration of advanced technologies such as whole exome sequencing (WES) and circulating tumor DNA (ctDNA) analysis plays a crucial role in monitoring treatment responses and identifying mechanisms of resistance [21]. By analyzing tumor samples and ctDNA, clinicians can make informed decisions regarding therapy adjustments, ensuring that patients receive the most effective treatments based on their current disease status.
The combination of targeted therapy with immunotherapy is another emerging strategy in melanoma treatment. Studies have shown that combining BRAF inhibitors with immune checkpoint inhibitors (such as PD-1 blockers) can lead to enhanced therapeutic responses compared to monotherapy [23]. This combinatorial approach aims to harness the strengths of both modalities—targeted therapy's rapid and potent action against specific tumor cells and immunotherapy's potential for durable responses by engaging the immune system.
Overall, the clinical applications of targeted therapies in melanoma underscore the shift towards precision medicine, where treatments are tailored to the individual patient's tumor characteristics. This paradigm not only improves therapeutic efficacy but also aims to minimize unnecessary toxicity associated with conventional therapies, ultimately leading to better patient outcomes and quality of life [24].
4 Challenges and Limitations
4.1 Tumor Heterogeneity
Targeted therapy in precision oncology aims to customize treatment based on the unique genetic, molecular, and phenotypic characteristics of an individual patient's tumor. This approach recognizes the inherent complexity and heterogeneity of cancer, which is characterized by significant variations both between different patients (intertumor heterogeneity) and within the same tumor (intratumor heterogeneity) [2][25][26].
The efficacy of targeted therapies is contingent upon the presence of specific molecular targets that are often identified through genomic profiling of tumors. These therapies are designed to inhibit particular pathways or molecules that are critical for cancer cell survival and proliferation. However, tumor heterogeneity poses a significant challenge to the effectiveness of these therapies. Heterogeneous tumors may harbor a diverse array of genetic mutations and phenotypic traits, which can lead to varying responses to treatment among different cell populations within the same tumor [27][28].
One of the primary limitations associated with targeted therapy is the potential for treatment resistance. As tumors evolve, they can develop new mutations that render previously effective therapies ineffective. This phenomenon is exacerbated by the presence of subpopulations of cancer cells that may inherently resist certain treatments [26][29]. The emergence of these resistant clones can lead to treatment failure and disease progression, underscoring the necessity for ongoing monitoring and adaptation of therapeutic strategies [4][30].
Moreover, the complexity of the tumor microenvironment, which includes interactions between cancer cells and surrounding stromal cells, further complicates the treatment landscape. This microenvironment can influence drug delivery and efficacy, making it essential to consider not only the cancer cells themselves but also the broader context in which they exist [26][31].
Despite the advancements in precision oncology, challenges such as high costs, limited accessibility, and ethical concerns regarding genetic data privacy remain significant barriers to the widespread implementation of targeted therapies [2][26]. Addressing these challenges requires continued research and innovation, particularly in developing novel strategies that can effectively target the diverse cellular populations within heterogeneous tumors [2][30].
In conclusion, while targeted therapy represents a promising approach within precision oncology, its effectiveness is significantly impacted by tumor heterogeneity, necessitating ongoing efforts to refine treatment modalities and develop strategies that can address the complexities of cancer biology.
4.2 Resistance Mechanisms
Targeted therapy in precision oncology operates by specifically inhibiting molecular targets that drive cancer cell proliferation and survival. This approach aims to personalize treatment based on the genetic and phenotypic characteristics of a patient's tumor, thereby enhancing therapeutic efficacy while minimizing toxicity. However, despite the advancements in targeted therapies, several challenges and limitations arise, particularly concerning resistance mechanisms.
Resistance to targeted therapies is a significant hurdle in the treatment of cancer. Cancer cells exhibit a remarkable ability to adapt and evolve, leading to intrinsic or acquired resistance that undermines the effectiveness of these therapies. The mechanisms of resistance can be broadly categorized into several classes.
Firstly, mutations in the target genes can render the therapy ineffective. For instance, alterations in the genes encoding for receptors or enzymes that targeted therapies aim to inhibit can prevent the drugs from binding effectively, thereby diminishing their therapeutic impact [32]. Secondly, the reactivation of previously inhibited signaling pathways can occur, allowing cancer cells to bypass the effects of the therapy. This phenomenon is often observed when cancer cells exploit alternative pathways to sustain their growth and survival [33].
Additionally, tumor microenvironment (TME) factors play a crucial role in the development of resistance. The TME can influence drug metabolism and efflux, leading to reduced drug efficacy. Changes in the TME, such as extracellular matrix remodeling or alterations in immune cell populations, can further complicate treatment responses [34]. Moreover, the intra-tumor heterogeneity of cancer cells often results in a diverse population of cells with varying susceptibilities to targeted therapies. This heterogeneity can limit the overall effectiveness of treatment, as some cells may survive and contribute to disease relapse [35].
Another layer of complexity arises from the plasticity of tumor cells, which can rapidly adapt to therapeutic pressures. For example, stress signaling pathways may be activated in response to targeted therapies, promoting cell survival and facilitating the emergence of resistant clones [36].
Emerging strategies to overcome these resistance mechanisms include combination therapies that target multiple pathways simultaneously, thereby reducing the likelihood of cancer cells developing resistance. Furthermore, novel approaches such as exploiting synthetic lethality and employing nanotechnology-based delivery systems are being explored to enhance the efficacy of targeted therapies [37].
In conclusion, while targeted therapy represents a significant advancement in precision oncology, its effectiveness is often compromised by various resistance mechanisms. A nuanced understanding of these mechanisms is essential for developing more effective treatment strategies that can adapt to the dynamic nature of cancer evolution and resistance. Continued research is necessary to elucidate these complex interactions and to innovate therapeutic approaches that can overcome resistance and improve patient outcomes.
4.3 Access and Affordability
Targeted therapy in precision oncology operates by utilizing molecular and genetic information about an individual’s tumor to tailor treatment strategies that specifically inhibit cancer growth and progression. This approach is founded on the premise that cancer is not a uniform disease but rather a collection of heterogeneous conditions driven by distinct molecular alterations. By identifying specific oncogenic drivers—such as mutations, amplifications, or translocations—clinicians can select targeted therapies that are designed to interact with these molecular targets, thereby optimizing therapeutic efficacy while minimizing damage to normal cells [26][38].
Despite the potential benefits of targeted therapies, several challenges and limitations impede their widespread implementation in clinical practice. One of the primary obstacles is the issue of access and affordability. The high costs associated with targeted therapies can be prohibitive for many patients and healthcare systems. These therapies often come with substantial price tags due to the extensive research and development processes required to bring them to market, coupled with the costs of companion diagnostics necessary for patient selection [39][40].
Access to these therapies is further complicated by disparities in healthcare systems, particularly in low-resource settings where advanced molecular testing may not be available. Additionally, reimbursement policies vary significantly, which can limit patient access to potentially life-saving treatments. Inconsistent coverage for molecular diagnostics and targeted therapies can lead to situations where patients do not receive appropriate treatment due to financial constraints or insurance limitations [13][41].
Furthermore, the dynamic nature of cancer, characterized by tumor heterogeneity and the potential for treatment resistance, poses additional challenges. Even with targeted therapies, patients may experience varying degrees of response, and some may develop resistance over time, necessitating ongoing research and adaptation of treatment strategies [5][40].
In summary, while targeted therapy in precision oncology offers a promising avenue for personalized cancer treatment, significant challenges related to access, affordability, and the inherent complexities of cancer biology must be addressed to realize its full potential in improving patient outcomes. Future efforts should focus on developing more inclusive healthcare policies, enhancing the affordability of targeted therapies, and ensuring equitable access to advanced diagnostic tools [2][42].
5 Future Directions in Targeted Therapy
5.1 Novel Target Identification
Targeted therapy in precision oncology operates on the principle of tailoring treatment based on the unique molecular characteristics of an individual's tumor. This approach leverages the understanding of specific genetic mutations and alterations within cancer cells, allowing for the development of therapies that directly target these anomalies. As the landscape of cancer treatment evolves, novel target identification has become a pivotal focus, driving advancements in therapeutic strategies.
Recent advancements in genomics and functional genetics have facilitated the unbiased discovery of new molecular targets, which is essential for the development of effective targeted therapies. For instance, the integration of genomic and functional genetic landscapes has shown promise in establishing a robust pipeline for cancer therapeutic targets, enabling the identification of actionable mutations that can be exploited for treatment (Konda et al., 2023) [43]. This approach aims to match patients with therapies that specifically target the molecular features of their tumors, thereby enhancing treatment efficacy and minimizing unnecessary side effects.
Moreover, precision oncology emphasizes the heterogeneity of cancer, recognizing that each patient's tumor may exhibit distinct genetic variations. This understanding underpins the rationale for personalized treatment plans, as the same therapeutic agent may yield different outcomes based on the genetic makeup of the tumor (Beecher et al., 2025) [2]. As a result, the development of targeted therapies has progressed from a one-size-fits-all model to a more individualized approach that considers the unique molecular profile of each patient's cancer.
The identification of disease-relevant genes and the development of molecularly targeted drugs have been greatly enhanced by advancements in technology, such as next-generation sequencing (NGS). NGS allows for the rapid and comprehensive analysis of genetic alterations, thereby facilitating the identification of potential therapeutic targets (Shin et al., 2017) [44]. This technology not only aids in the discovery of new targets but also assists in the validation of existing ones, ensuring that therapies are developed based on sound scientific evidence.
In terms of future directions, there is a strong emphasis on refining the methods used for target identification and validation. This includes exploring novel biomarkers that can predict patient responses to specific therapies and developing more sophisticated bioinformatics tools to analyze complex genomic data (Chae et al., 2017) [19]. The continued evolution of precision oncology will likely involve integrating multidisciplinary approaches, combining insights from genomics, proteomics, and patient clinical data to enhance the identification of actionable targets.
In conclusion, targeted therapy in precision oncology represents a significant shift towards personalized medicine, where treatment is tailored to the individual characteristics of each patient's tumor. The ongoing efforts in novel target identification, driven by advancements in genomic technologies and a deeper understanding of cancer biology, hold the potential to revolutionize cancer treatment and improve patient outcomes.
5.2 Combination Therapies
Targeted therapy in precision oncology represents a transformative approach to cancer treatment, characterized by its focus on specific molecular targets that drive tumor growth and progression. This methodology leverages the unique genetic, molecular, and clinical profiles of individual tumors to optimize therapeutic efficacy while minimizing adverse effects.
The core principle of targeted therapy is to inhibit key molecular pathways or genetic alterations that are essential for cancer cell survival and proliferation. These therapies include monoclonal antibodies and small molecule inhibitors that are designed to specifically interact with their targets, such as mutated proteins or aberrant signaling pathways. The advantages of targeted therapies over traditional chemotherapy are evident in their improved tolerability and specificity, which often result in better overall survival (OS) and progression-free survival (PFS) rates for patients receiving matched therapies [45].
As the field of precision oncology evolves, there is an increasing recognition of the potential of combination therapies. Combining targeted therapies with other treatment modalities, such as immunotherapy or radiotherapy, is emerging as a promising strategy to enhance treatment outcomes, particularly for patients with complex and heterogeneous tumors. The rationale behind combination therapies lies in their ability to address multiple pathways involved in tumorigenesis, thereby reducing the likelihood of resistance that often arises when targeting a single molecular alteration [46].
Recent literature underscores the importance of developing robust frameworks for the implementation of combination therapies in clinical practice. For instance, the integration of genomic profiling and molecular diagnostics can help identify actionable targets and facilitate the selection of appropriate combination regimens. Studies indicate that personalized treatment strategies, which utilize comprehensive molecular profiling to inform the choice of drug combinations, may lead to significant improvements in disease control and patient survival [4].
Moreover, the incorporation of innovative approaches such as antibody-oligonucleotide conjugates (AOCs) is being explored as a means to enhance the specificity and efficacy of targeted therapies. These conjugates aim to deliver therapeutic agents directly to cancer cells, potentially overcoming some of the limitations associated with traditional targeted therapies [8].
The future of targeted therapy in precision oncology is likely to be shaped by advancements in genomics and bioinformatics, which will facilitate the identification of novel molecular targets and the development of combination strategies that are tailored to the individual patient's tumor profile. Ongoing clinical trials and research efforts will be crucial in determining the most effective combinations and sequencing strategies to optimize treatment outcomes [43].
In conclusion, targeted therapy in precision oncology is a dynamic and rapidly evolving field. The integration of combination therapies holds significant promise for improving the management of cancer, particularly as our understanding of tumor biology and the molecular mechanisms underlying cancer continues to deepen. As researchers and clinicians strive to refine these approaches, the goal remains to provide personalized, effective, and safe treatment options for all cancer patients.
5.3 Personalized Medicine Approaches
Targeted therapy in precision oncology operates by utilizing specific molecular or genetic alterations within a patient's tumor to tailor treatment strategies that are most likely to be effective for that individual. This approach acknowledges the inherent heterogeneity of cancer, where even tumors of the same type can exhibit distinct genetic profiles and responses to treatment. The primary goal is to optimize therapeutic efficacy while minimizing adverse effects.
The process begins with comprehensive genomic profiling of the tumor, which identifies actionable mutations or alterations that can be targeted by specific therapies. For instance, certain targeted therapies are designed to inhibit key molecular pathways or mechanisms that promote tumor growth and survival. This strategy has evolved significantly over the past two decades, with numerous clinical trials demonstrating the potential benefits of targeted therapies. However, the implementation of these therapies is often challenged by the complexity of cancer biology and the emergence of resistance mechanisms[5].
One of the promising advancements in targeted therapy is the development of organelle-targeted therapies. These strategies focus on delivering therapeutic agents directly to specific organelles within cancer cells, such as mitochondria, lysosomes, or the endoplasmic reticulum. By concentrating the drug's action within these cellular compartments, it is possible to enhance therapeutic efficacy and reduce systemic toxicity, which is a common limitation of traditional cancer treatments[13].
Moreover, precision oncology is not limited to genomic profiling alone. The incorporation of functional precision medicine, which tests live tumor cells with various drugs to identify vulnerabilities, represents a significant evolution in the field. This method allows for immediate, personalized treatment recommendations based on the tumor's specific response to drugs, rather than relying solely on static genetic information[11].
As the field progresses, the integration of bioinformatics tools and data platforms will be crucial for improving the actionability of molecular lesions and supporting patient management. These tools can help to standardize frameworks for implementing targeted therapies in clinical settings, thereby enhancing patient outcomes[19].
Looking towards the future, personalized medicine approaches in targeted therapy will likely expand further. This includes the exploration of combination therapies that address multiple molecular targets simultaneously, potentially improving response rates in patients with complex and heterogeneous cancers. Current research emphasizes the importance of not only identifying driver mutations but also considering the dynamic nature of tumor evolution, which may necessitate adaptive treatment strategies over time[47].
In conclusion, targeted therapy in precision oncology represents a paradigm shift towards individualized treatment strategies that are informed by the unique genetic and molecular characteristics of a patient's tumor. The future directions in this field will likely involve more sophisticated integration of genomic data, functional assessments, and novel therapeutic modalities to enhance the effectiveness and precision of cancer treatments.
6 Conclusion
Targeted therapy in precision oncology has emerged as a transformative approach in cancer treatment, leveraging the unique molecular and genetic characteristics of individual tumors to optimize therapeutic efficacy while minimizing adverse effects. This report highlights several key findings regarding the mechanisms, clinical applications, and challenges associated with targeted therapies. The mechanisms of targeted therapy include the inhibition of specific signaling pathways, antibody-drug conjugates, immune checkpoint inhibition, and the use of personalized biomarkers to guide treatment decisions. The clinical applications of targeted therapies in breast cancer, lung cancer, and melanoma demonstrate the potential for improved patient outcomes through tailored treatment strategies. However, challenges such as tumor heterogeneity, resistance mechanisms, and access to therapies remain significant barriers to the widespread implementation of targeted therapies. Future research directions should focus on novel target identification, combination therapies, and personalized medicine approaches that address the complexities of cancer biology. By overcoming these challenges and advancing our understanding of cancer mechanisms, targeted therapy can continue to evolve, ultimately leading to better patient outcomes and a more effective management of cancer.
References
- [1] Won Duk Joo;Irene Visintin;Gil Mor. Targeted cancer therapy--are the days of systemic chemotherapy numbered?. Maturitas(IF=3.6). 2013. PMID:24128673. DOI: .
- [2] Kate Beecher;Tivya Kulasegaran;Sunil R Lakhani;Amy E McCart Reed. Genomic Predictive Biomarkers in Breast Cancer: The Haves and Have Nots.. International journal of molecular sciences(IF=4.9). 2025. PMID:40806432. DOI: 10.3390/ijms26157300.
- [3] Md Abdus Subhan;Farzana Parveen;Hassan Shah;Satya Siva Kishan Yalamarty;Janaína Artem Ataide;Valdimir P Torchilin. Recent Advances with Precision Medicine Treatment for Breast Cancer including Triple-Negative Sub-Type.. Cancers(IF=4.4). 2023. PMID:37190133. DOI: 10.3390/cancers15082204.
- [4] Jason K Sicklick;Shumei Kato;Ryosuke Okamura;Maria Schwaederle;Michael E Hahn;Casey B Williams;Pradip De;Amy Krie;David E Piccioni;Vincent A Miller;Jeffrey S Ross;Adam Benson;Jennifer Webster;Philip J Stephens;J Jack Lee;Paul T Fanta;Scott M Lippman;Brian Leyland-Jones;Razelle Kurzrock. Molecular profiling of cancer patients enables personalized combination therapy: the I-PREDICT study.. Nature medicine(IF=50.0). 2019. PMID:31011206. DOI: 10.1038/s41591-019-0407-5.
- [5] Andrea Nicolini;Paola Ferrari. Targeted Therapies and Drug Resistance in Advanced Breast Cancer, Alternative Strategies and the Way beyond.. Cancers(IF=4.4). 2024. PMID:38275906. DOI: 10.3390/cancers16020466.
- [6] Robert G Bristow;Brian Alexander;Michael Baumann;Scott V Bratman;J Martin Brown;Kevin Camphausen;Peter Choyke;Deborah Citrin;Joseph N Contessa;Adam Dicker;David G Kirsch;Mechthild Krause;Quynh-Thu Le;Michael Milosevic;Zachary S Morris;Jann N Sarkaria;Paul M Sondel;Phuoc T Tran;George D Wilson;Henning Willers;Rebecca K S Wong;Paul M Harari. Combining precision radiotherapy with molecular targeting and immunomodulatory agents: a guideline by the American Society for Radiation Oncology.. The Lancet. Oncology(IF=35.9). 2018. PMID:29726389. DOI: 10.1016/S1470-2045(18)30096-2.
- [7] Suman Paul;Maximilian F Konig;Drew M Pardoll;Chetan Bettegowda;Nickolas Papadopoulos;Katharine M Wright;Sandra B Gabelli;Mitchell Ho;Andrea van Elsas;Shibin Zhou. Cancer therapy with antibodies.. Nature reviews. Cancer(IF=66.8). 2024. PMID:38740967. DOI: 10.1038/s41568-024-00690-x.
- [8] Jinlan Jiao;Yun Qian;Yinhua Lv;Wenqian Wei;Yongxuan Long;Xiaoling Guo;Anya Buerliesi;Jiahui Ye;Hao Han;Jinbo Li;Yun Zhu;Weijie Zhang. Overcoming limitations and advancing the therapeutic potential of antibody-oligonucleotide conjugates (AOCs): Current status and future perspectives.. Pharmacological research(IF=10.5). 2024. PMID:39433169. DOI: 10.1016/j.phrs.2024.107469.
- [9] Luo Wang;Jingao Li;Xuanwei Zeng;Qinyue Lu;Yifeng Wang;Zulhumar Anwar;Li Xue;Heli Fan;Huabing Sun. Exogenous/endogenous stimuli-responsive antitumor prodrugs advance precision chemotherapy.. Bioorganic & medicinal chemistry(IF=3.0). 2025. PMID:41253066. DOI: 10.1016/j.bmc.2025.118485.
- [10] Xing Wang;Yuqi Tang;Quan Li. Mitochondria-Targeted Molecular Tools in Precise Tumor Therapy.. Angewandte Chemie (International ed. in English)(IF=16.9). 2025. PMID:40790919. DOI: 10.1002/anie.202511230.
- [11] Anthony Letai;Patrick Bhola;Alana L Welm. Functional precision oncology: Testing tumors with drugs to identify vulnerabilities and novel combinations.. Cancer cell(IF=44.5). 2022. PMID:34951956. DOI: 10.1016/j.ccell.2021.12.004.
- [12] Sara S Bashraheel;Alexander Domling;Sayed K Goda. Update on targeted cancer therapies, single or in combination, and their fine tuning for precision medicine.. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie(IF=7.5). 2020. PMID:32106381. DOI: 10.1016/j.biopha.2020.110009.
- [13] Jingjing Yang;Anthony Griffin;Zhe Qiang;Jie Ren. Organelle-targeted therapies: a comprehensive review on system design for enabling precision oncology.. Signal transduction and targeted therapy(IF=52.7). 2022. PMID:36402753. DOI: 10.1038/s41392-022-01243-0.
- [14] Jianzhi Zhang;Hao Zhu;Wentao Liu;Ji Miao;Yonghuan Mao;Qiang Li. Prognostic and predictive molecular biomarkers in colorectal cancer.. Frontiers in oncology(IF=3.3). 2025. PMID:40308511. DOI: 10.3389/fonc.2025.1532924.
- [15] Shriyansh Srivastava;Amit Anand;Anil Goud Kandhula;Pooja Jaiswal;Sachin Kumar;Monica Gulati;Monika Sachdeva;Tapan Behl. Precision Medicine in Colorectal Cancer: Targeted Therapies and Biomarker Insights.. Current cancer drug targets(IF=3.5). 2025. PMID:39878113. DOI: 10.2174/0115680096338273241224061021.
- [16] Soo-Ryum Yang;Anne M Schultheis;Helena Yu;Diana Mandelker;Marc Ladanyi;Reinhard Büttner. Precision medicine in non-small cell lung cancer: Current applications and future directions.. Seminars in cancer biology(IF=15.7). 2022. PMID:32730814. DOI: 10.1016/j.semcancer.2020.07.009.
- [17] Christopher E Barbieri;Arul M Chinnaiyan;Seth P Lerner;Charles Swanton;Mark A Rubin. The Emergence of Precision Urologic Oncology: A Collaborative Review on Biomarker-driven Therapeutics.. European urology(IF=25.2). 2017. PMID:27567210. DOI: 10.1016/j.eururo.2016.08.024.
- [18] Dae Ho Lee. Practical issues of biomarker-assisted targeted therapy in precision medicine and immuno-oncology era.. ESMO open(IF=8.3). 2018. PMID:29942670. DOI: 10.1136/esmoopen-2018-000370.
- [19] Young Kwang Chae;Alan P Pan;Andrew A Davis;Sandip P Patel;Benedito A Carneiro;Razelle Kurzrock;Francis J Giles. Path toward Precision Oncology: Review of Targeted Therapy Studies and Tools to Aid in Defining "Actionability" of a Molecular Lesion and Patient Management Support.. Molecular cancer therapeutics(IF=5.5). 2017. PMID:29203694. DOI: 10.1158/1535-7163.MCT-17-0597.
- [20] Keith T Flaherty;Grant McArthur. BRAF, a target in melanoma: implications for solid tumor drug development.. Cancer(IF=5.1). 2010. PMID:20629085. DOI: 10.1002/cncr.25261.
- [21] Maria Romina Girotti;Gabriela Gremel;Rebecca Lee;Elena Galvani;Dominic Rothwell;Amaya Viros;Amit Kumar Mandal;Kok Haw Jonathan Lim;Grazia Saturno;Simon J Furney;Franziska Baenke;Malin Pedersen;Jane Rogan;Jacqueline Swan;Matthew Smith;Alberto Fusi;Deemesh Oudit;Nathalie Dhomen;Ged Brady;Paul Lorigan;Caroline Dive;Richard Marais. Application of Sequencing, Liquid Biopsies, and Patient-Derived Xenografts for Personalized Medicine in Melanoma.. Cancer discovery(IF=33.3). 2016. PMID:26715644. DOI: 10.1158/2159-8290.CD-15-1336.
- [22] Markus Reitmajer;Lukas Flatz. Precision medicine in cutaneous melanoma-A comprehensive review.. Journal of the European Academy of Dermatology and Venereology : JEADV(IF=8.0). 2025. PMID:41081438. DOI: 10.1111/jdv.70111.
- [23] Jennifer A Wargo;Zachary A Cooper;Keith T Flaherty. Universes collide: combining immunotherapy with targeted therapy for cancer.. Cancer discovery(IF=33.3). 2014. PMID:25395294. DOI: 10.1158/2159-8290.CD-14-0477.
- [24] Paolo A Ascierto;Christian Blank;Reinhard Dummer;Marc S Ernstoff;Soldano Ferrone;Bernard A Fox;Thomas F Gajewski;Claus Garbe;Patrick Hwu;Pawel Kalinski;Michelle Krogsgaard;Roger S Lo;Jason J Luke;Bart Neyns;Michael A Postow;Sergio A Quezada;Michele W L Teng;Giorgio Trinchieri;Alessandro Testori;Corrado Caracò;Iman Osman;Igor Puzanov;Magdalena Thurin. Perspectives in Melanoma: meeting report from the Melanoma Bridge (December 3rd-5th, 2020, Italy).. Journal of translational medicine(IF=7.5). 2021. PMID:34193182. DOI: 10.1186/s12967-021-02951-x.
- [25] María José Jiménez-Santos;Santiago García-Martín;Coral Fustero-Torre;Tomás Di Domenico;Gonzalo Gómez-López;Fátima Al-Shahrour. Bioinformatics roadmap for therapy selection in cancer genomics.. Molecular oncology(IF=4.5). 2022. PMID:35811332. DOI: 10.1002/1878-0261.13286.
- [26] Rituraj;Rashmi Saxena Pal;Jenny Wahlang;Yogendra Pal;Mvnl Chaitanya;Sumedha Saxena. Precision oncology: transforming cancer care through personalized medicine.. Medical oncology (Northwood, London, England)(IF=3.5). 2025. PMID:40488843. DOI: 10.1007/s12032-025-02817-y.
- [27] Mariona Baliu-Piqué;Atanasio Pandiella;Alberto Ocana. Breast Cancer Heterogeneity and Response to Novel Therapeutics.. Cancers(IF=4.4). 2020. PMID:33167363. DOI: 10.3390/cancers12113271.
- [28] Duojiao Wu;Diane C Wang;Yunfeng Cheng;Mengjia Qian;Miaomiao Zhang;Qi Shen;Xiangdong Wang. Roles of tumor heterogeneity in the development of drug resistance: A call for precision therapy.. Seminars in cancer biology(IF=15.7). 2017. PMID:27840278. DOI: 10.1016/j.semcancer.2016.11.006.
- [29] Jialing Zhang;Stephan Stanislaw Späth;Sadie L Marjani;Wengeng Zhang;Xinghua Pan. Characterization of cancer genomic heterogeneity by next-generation sequencing advances precision medicine in cancer treatment.. Precision clinical medicine(IF=5.0). 2018. PMID:30687561. DOI: 10.1093/pcmedi/pby007.
- [30] Aleksandr Ianevski;Kristen Nader;Kyriaki Driva;Wojciech Senkowski;Daria Bulanova;Lidia Moyano-Galceran;Tanja Ruokoranta;Heikki Kuusanmäki;Nemo Ikonen;Philipp Sergeev;Markus Vähä-Koskela;Anil K Giri;Anna Vähärautio;Mika Kontro;Kimmo Porkka;Esa Pitkänen;Caroline A Heckman;Krister Wennerberg;Tero Aittokallio. Single-cell transcriptomes identify patient-tailored therapies for selective co-inhibition of cancer clones.. Nature communications(IF=15.7). 2024. PMID:39362905. DOI: 10.1038/s41467-024-52980-5.
- [31] Dilip R Gunturu;Mohammed Hassan;Deepa Bedi;Pran Datta;Upender Manne;Temesgen Samuel. Unlocking the Potential of Therapy-Induced Cytokine Responses: Illuminating New Pathways in Cancer Precision Medicine.. Current oncology (Toronto, Ont.)(IF=3.4). 2024. PMID:38534922. DOI: 10.3390/curroncol31030089.
- [32] P Ramos;M Bentires-Alj. Mechanism-based cancer therapy: resistance to therapy, therapy for resistance.. Oncogene(IF=7.3). 2015. PMID:25263438. DOI: 10.1038/onc.2014.314.
- [33] Peter T Harrison;Paul H Huang. Exploiting vulnerabilities in cancer signalling networks to combat targeted therapy resistance.. Essays in biochemistry(IF=5.7). 2018. PMID:30072489. DOI: 10.1042/EBC20180016.
- [34] Nolan M Stubbs;Tyler J Roady;Maximilian P Schwermann;Elias O Eteshola;William J MacDonald;Connor Purcell;Dinara Ryspayeva;Nataliia Verovkina;Vida Tajiknia;Maryam Ghandali;Viva Voong;Alexis J Lannigan;Alexander G Raufi;Sean Lawler;Sheldon L Holder;Benedito A Carneiro;Liang Cheng;Howard P Safran;Stephanie L Graff;Don S Dizon;Sendurai A Mani;Attila A Seyhan;Robert W Sobol;Eric T Wong;Clark C Chen;Ziya Gokaslan;Martin S Taylor;Brian M Rivers;Wafik S El-Deiry. Acquired resistance to molecularly targeted therapies for cancer.. Cancer drug resistance (Alhambra, Calif.)(IF=5.2). 2025. PMID:40510029. DOI: 10.20517/cdr.2024.189.
- [35] Catherine Vander Linden;Cyril Corbet. Reconciling environment-mediated metabolic heterogeneity with the oncogene-driven cancer paradigm in precision oncology.. Seminars in cell & developmental biology(IF=6.0). 2020. PMID:31103464. DOI: 10.1016/j.semcdb.2019.05.016.
- [36] E Pazarentzos;T G Bivona. Adaptive stress signaling in targeted cancer therapy resistance.. Oncogene(IF=7.3). 2015. PMID:25703329. DOI: 10.1038/onc.2015.26.
- [37] Yan Gao;Jacson K Shen;Lara Milane;Francis J Hornicek;Mansoor M Amiji;Zhenfeng Duan. Targeted cancer therapy; nanotechnology approaches for overcoming drug resistance.. Current medicinal chemistry(IF=3.5). 2015. PMID:25666804. DOI: 10.2174/0929867322666150209151851.
- [38] Joel Rivera-Concepcion;Dipesh Uprety;Alex A Adjei. Challenges in the Use of Targeted Therapies in Non-Small Cell Lung Cancer.. Cancer research and treatment(IF=3.8). 2022. PMID:35209703. DOI: 10.4143/crt.2022.078.
- [39] D Dankó;J-Y Blay;L P Garrison. Challenges in the value assessment, pricing and funding of targeted combination therapies in oncology.. Health policy (Amsterdam, Netherlands)(IF=3.4). 2019. PMID:31337514. DOI: 10.1016/j.healthpol.2019.07.009.
- [40] Hassan Maleki;Hammed Tanimowo Aiyelabegan;Parisa Javadi;Fereshteh Abdi;Farshad Mirzavi;Zeinab Zarei Behjani;Albert A Rizvanov;Sara Takallu;Rajesh Kumar;Reyhaneh Hadi Barhaghtalab;Arghavan Hosseinpouri;Roghayyeh Vakili-Ghartavol. Nanotechnology-mediated precision drug delivery strategies for breast cancer treatment.. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie(IF=7.5). 2025. PMID:40456194. DOI: 10.1016/j.biopha.2025.118224.
- [41] Lincoln D Nadauld;James M Ford;Daryl Pritchard;Thomas Brown. Strategies For Clinical Implementation: Precision Oncology At Three Distinct Institutions.. Health affairs (Project Hope)(IF=8.1). 2018. PMID:29733728. DOI: 10.1377/hlthaff.2017.1575.
- [42] Jakob M Riedl;Florian Moik;Tamara Esterl;Sarah M Kostmann;Armin Gerger;Philipp J Jost. Molecular diagnostics tailoring personalized cancer therapy-an oncologist's view.. Virchows Archiv : an international journal of pathology(IF=3.1). 2024. PMID:37982847. DOI: 10.1007/s00428-023-03702-7.
- [43] Prathyusha Konda;Simon Garinet;Eliezer M Van Allen;Srinivas R Viswanathan. Genome-guided discovery of cancer therapeutic targets.. Cell reports(IF=6.9). 2023. PMID:37572322. DOI: 10.1016/j.celrep.2023.112978.
- [44] Seung Ho Shin;Ann M Bode;Zigang Dong. Addressing the challenges of applying precision oncology.. NPJ precision oncology(IF=8.0). 2017. PMID:29872710. DOI: 10.1038/s41698-017-0032-z.
- [45] Zhijun Zhou;Min Li. Targeted therapies for cancer.. BMC medicine(IF=8.3). 2022. PMID:35272686. DOI: 10.1186/s12916-022-02287-3.
- [46] Sarah Bowen Jones;Clara Chan;Andrea R Filippi;Ken Harada;Alexander V Louie;Colin R Lindsay;Ernest Nadal;Pablo Munoz Schuffenegger;David Woolf;Corinne Faivre-Finn; . Emerging Role of Targeted Therapies Combined With Radiotherapy in Inoperable Stages I to III NSCLC: A Review From the IASLC ART Subcommittee.. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer(IF=20.8). 2025. PMID:40350081. DOI: 10.1016/j.jtho.2025.05.004.
- [47] Nabil Amirouchene-Angelozzi;Charles Swanton;Alberto Bardelli. Tumor Evolution as a Therapeutic Target.. Cancer discovery(IF=33.3). 2017. PMID:28729406. DOI: 10.1158/2159-8290.CD-17-0343.
MaltSci Intelligent Research Services
Search for more papers on MaltSci.com
Targeted Therapy · Precision Oncology · Biomarkers · Tumor Heterogeneity · Personalized Medicine
© 2025 MaltSci
