Appearance
This report is written by MaltSci based on the latest literature and research findings
What are the latest drug targets for cancer?
Abstract
Cancer remains a major global health challenge, necessitating the ongoing exploration of novel drug targets to improve treatment outcomes. Recent advancements in molecular biology, particularly genomics and proteomics, have facilitated the identification of new drug targets that promise to enhance cancer therapies' efficacy while minimizing adverse effects. This review provides a comprehensive overview of the latest drug targets in cancer therapy, focusing on oncogenes, tumor suppressor genes, immune checkpoints, and novel signaling pathways. Targeted therapies have transformed the treatment landscape, moving towards precise interventions that disrupt the molecular underpinnings of cancer. Significant progress has been made with small-molecule inhibitors, monoclonal antibodies, and CAR-T cell therapies, demonstrating their clinical relevance and potential to reshape therapeutic approaches. Current clinical trials are evaluating emerging drug targets, revealing promising options for patients with previously limited therapeutic alternatives. However, challenges such as drug resistance and the complexity of tumor biology remain significant hurdles. Future directions in cancer therapy will likely emphasize personalized medicine, aiming to tailor treatments based on individual tumor profiles, alongside the integration of combination therapies to enhance efficacy. This synthesis of recent findings aims to elucidate the dynamic field of cancer drug development, providing insights into the future of targeted therapies and their role in improving patient outcomes.
Outline
This report will discuss the following questions.
- 1 Introduction
- 2 Overview of Cancer Drug Targets
- 2.1 Oncogenes as Drug Targets
- 2.2 Tumor Suppressor Genes and Their Therapeutic Implications
- 2.3 Immune Checkpoints and Immunotherapy
- 2.4 Novel Signaling Pathways in Cancer
- 3 Recent Advances in Targeted Therapies
- 3.1 Small Molecule Inhibitors
- 3.2 Monoclonal Antibodies
- 3.3 CAR-T Cell Therapy
- 4 Clinical Trials and Emerging Drug Targets
- 4.1 Overview of Current Clinical Trials
- 4.2 Promising New Targets in Development
- 5 Challenges and Future Directions
- 5.1 Resistance Mechanisms
- 5.2 Personalized Medicine Approaches
- 5.3 Combination Therapies
- 6 Summary
1 Introduction
Cancer remains a significant global health challenge, being one of the leading causes of morbidity and mortality. According to the World Health Organization, cancer accounted for approximately 10 million deaths in 2020 alone, underscoring the urgent need for innovative therapeutic strategies [1]. The complexity of cancer biology, characterized by genetic heterogeneity and adaptive resistance mechanisms, necessitates the ongoing exploration of novel drug targets to improve treatment outcomes across various cancer types [2]. Recent advancements in molecular biology, particularly the integration of genomics and proteomics, have facilitated the identification of new drug targets that promise to enhance the efficacy of cancer therapies while minimizing adverse effects [3].
The significance of identifying and targeting specific molecular pathways in cancer cannot be overstated. Targeted therapies have transformed the treatment landscape, moving away from traditional cytotoxic agents towards more precise interventions that aim to disrupt the molecular underpinnings of cancer [4]. The development of agents targeting oncogenes, tumor suppressor genes, immune checkpoints, and novel signaling pathways has opened new avenues for therapeutic intervention [5]. Moreover, the advent of personalized medicine, which tailors treatment based on individual tumor profiles, represents a paradigm shift in oncology, aiming to enhance treatment specificity and efficacy [6].
Current research highlights a plethora of drug targets, including receptor tyrosine kinases, which have been pivotal in the development of small-molecule inhibitors and monoclonal antibodies [1][2]. Notable examples include trastuzumab for HER2-positive breast cancer and imatinib for chronic myeloid leukemia, both of which have significantly improved patient outcomes [7]. Additionally, the exploration of immune checkpoint inhibitors has revolutionized cancer immunotherapy, providing new options for patients who previously had limited therapeutic alternatives [8].
This review will provide a comprehensive overview of the latest drug targets in cancer therapy, organized as follows:
Overview of Cancer Drug Targets: This section will discuss the role of oncogenes, tumor suppressor genes, immune checkpoints, and novel signaling pathways as potential drug targets. Each category will be explored in terms of its mechanisms of action and therapeutic implications.
Recent Advances in Targeted Therapies: We will examine the development of small-molecule inhibitors, monoclonal antibodies, and CAR-T cell therapies, emphasizing their clinical relevance and the latest findings from ongoing trials.
Clinical Trials and Emerging Drug Targets: An overview of current clinical trials will be presented, highlighting promising new targets under investigation, which could potentially reshape treatment paradigms.
Challenges and Future Directions: This section will address the challenges posed by drug resistance mechanisms, the role of personalized medicine, and the potential of combination therapies to enhance treatment efficacy.
Summary: Finally, we will summarize the key findings and discuss future directions for drug development in the context of the evolving landscape of cancer therapeutics.
Through this synthesis of recent findings and ongoing research efforts, this review aims to elucidate the dynamic field of cancer drug development, offering insights into the future of targeted therapies and their role in improving patient outcomes.
2 Overview of Cancer Drug Targets
2.1 Oncogenes as Drug Targets
The landscape of cancer treatment has evolved significantly over the years, particularly in the identification and validation of drug targets that are crucial for the development of effective therapies. The latest drug targets for cancer can be broadly categorized into several key areas, including oncogenes, tumor suppressor genes, and other molecular pathways.
Recent advancements have highlighted the importance of targeting specific oncogenes and their associated pathways. This is particularly relevant as the majority of cancers arise from multiple genetic lesions, necessitating the use of sophisticated drug cocktails or single agents that can act on multiple downstream targets to enhance therapeutic efficacy (Burger 2007). The emerging paradigm in cancer therapy is to develop agents that inhibit validated targets that are relatively specific for tumors, thus minimizing damage to normal cells (Burger 2007).
In addition to oncogenes, there is an increasing focus on proteins that are essential for cancer cell survival and adaptation to the unique stresses of the tumor microenvironment. These targets are not necessarily encoded by oncogenes but are crucial for the cancer phenotype (Moscat et al. 2015). This approach broadens the scope of potential therapeutic targets beyond traditional oncogenes and tumor suppressor genes.
Furthermore, molecularly targeted therapies have been integrated with cytotoxic and anti-endocrine drugs, emphasizing the need to target molecular pathways that sustain the carcinogenic process. Key molecular targets for anticancer drug development include cell surface receptors, signal transduction pathways, gene transcription targets, and components of the ubiquitin-proteasome system (Yamanaka & Saya 2009; Rosa et al. 2008). These strategies aim to disrupt critical processes that contribute to tumor growth and metastasis.
The advancements in genomic technologies have also played a pivotal role in identifying actionable alterations in oncogene-driven cancers, which has led to the development of precision oncology approaches (Zugazagoitia et al. 2016). However, challenges remain, particularly in the areas of tumor heterogeneity and acquired resistance, which complicate the effectiveness of targeted therapies.
In summary, the latest drug targets for cancer include not only oncogenes but also a wider array of molecular pathways that are essential for tumor survival and proliferation. The ongoing research and development in this field aim to create more effective, safe, and targeted therapies that can significantly improve patient outcomes.
2.2 Tumor Suppressor Genes and Their Therapeutic Implications
Recent advancements in cancer research have highlighted tumor suppressor genes (TSGs) as significant drug targets due to their critical roles in regulating cell growth and maintaining genomic integrity. The dysregulation of these genes can lead to cancer development and progression, making them attractive candidates for therapeutic interventions.
Tumor suppressor genes, such as p53, PTEN, and Rb, are frequently altered in various cancers, leading to unchecked cell proliferation and resistance to apoptosis. The restoration of TSG function is a promising strategy in cancer therapy. For instance, the wild-type p53 gene has been a focal point of gene therapy efforts, with clinical trials demonstrating its potential to enhance the efficacy of conventional chemotherapy and radiotherapy by sensitizing tumors to these treatments [9].
Moreover, the identification of specific genomic loci associated with loss of heterozygosity (LOH) in TSGs has provided insights into their role in tumorigenesis. Research indicates that certain loci, such as 3p14.2 (FHIT), 9p21.3 (p16INK4a), 10q23 (PTEN), and 17p13 (TP53), are particularly susceptible to LOH due to environmental and genetic factors, including exposure to carcinogens and chromosomal instability [10]. This understanding of TSG alterations is essential for developing targeted therapies that can effectively restore TSG function in cancer cells.
The therapeutic implications of targeting TSGs extend beyond gene replacement strategies. For example, the use of small interfering RNA (siRNA) technology for tumor-directed delivery aims to silence the expression of oncogenes or restore the function of TSGs, offering a tailored approach to cancer treatment [11]. The success of such targeted therapies hinges on the development of effective delivery systems that minimize off-target effects and maximize therapeutic efficacy.
Furthermore, the interplay between TSGs and chemotherapeutic drug response has been extensively studied. TSGs not only play a role in tumor initiation and progression but also significantly influence how tumors respond to various chemotherapeutic agents. For instance, mutations in TSGs can lead to drug resistance, underscoring the importance of incorporating TSG status into treatment planning for better prediction of clinical outcomes [12].
In conclusion, tumor suppressor genes represent a critical area of focus in the development of novel cancer therapies. Their role in regulating cellular processes and their frequent alteration in cancer make them valuable targets for therapeutic strategies aimed at restoring normal cellular function and improving patient outcomes. Ongoing research into the molecular mechanisms underlying TSG dysregulation and the development of innovative delivery systems will further enhance the potential of TSG-targeted therapies in clinical settings.
2.3 Immune Checkpoints and Immunotherapy
Recent advancements in cancer therapy have emphasized the identification and targeting of various drug targets, particularly in the realm of immunotherapy. Immune checkpoints have emerged as critical targets in the development of novel cancer treatments. These checkpoints are immunosuppressive molecules that can inhibit immune activation, thereby allowing cancer cells to evade immune surveillance. The exploration of immune checkpoint inhibitors (ICIs) has revolutionized cancer therapy, leading to significant breakthroughs in patient outcomes.
Recent studies have highlighted several new immune checkpoint molecules that present promising therapeutic targets. These include lymphocyte activation gene-3 (LAG-3), B and T lymphocyte attenuator (BTLA), programmed death-1 homolog (PD-1H), T-cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIM-3)/carcinoembryonic antigen cell adhesion molecule 1 (CEACAM1), and the poliovirus receptor (PVR)-like receptors. Research into these molecules has yielded encouraging results in preclinical settings, indicating their potential as targets for enhancing cancer immunotherapy [13].
In addition to traditional immune checkpoints such as PD-1 and CTLA-4, the exploration of neoantigens has gained traction as a promising strategy in cancer immunotherapy. Neoantigens are mutated peptides unique to tumor cells that can elicit strong T-cell responses without affecting normal tissues. Advances in next-generation sequencing technologies facilitate the identification of these neoantigens, which can be targeted through personalized vaccines and adoptive T-cell therapies [14].
Furthermore, the integration of novel methodologies, including therapeutic vaccines, CAR-T cell therapies, and small molecule drugs, has marked a new era in cancer treatment. These approaches are designed to personalize cancer therapies based on the genetic and molecular profile of individual tumors. This shift towards personalized medicine aims to enhance the efficacy of treatments while minimizing toxicity [15].
The mechanisms of immune checkpoint inhibitors have also been a focal point of research. ICIs such as anti-PD-1 and anti-PD-L1 have shown remarkable efficacy across various cancer types, although their overall response rates remain modest, typically ranging from 10% to 30% in clinical settings. The primary and acquired resistance to these therapies presents significant challenges, necessitating a deeper understanding of the molecular and regulatory mechanisms governing immune checkpoints [16].
Recent trends indicate a growing interest in combination therapies that synergize the effects of ICIs with other therapeutic modalities, including chemotherapy and targeted therapies. This strategy aims to overcome resistance mechanisms and improve patient outcomes. Ongoing clinical trials are investigating the efficacy of these combination approaches across different cancer types [17].
In summary, the landscape of cancer drug targets is rapidly evolving, with immune checkpoints and neoantigens at the forefront of research and clinical application. The development of novel therapeutic strategies aimed at these targets holds great promise for improving the efficacy of cancer immunotherapy and achieving better clinical outcomes for patients.
2.4 Novel Signaling Pathways in Cancer
Recent advancements in cancer therapy have focused on novel signaling pathways and drug targets that offer new avenues for treatment. These developments are crucial as cancer remains a leading global health issue, necessitating innovative therapeutic strategies.
One of the most significant areas of research is the identification of molecular pathways implicated in cancer progression. Targeted therapies have emerged as a key strategy, directing treatment towards specific molecular alterations within tumors. For instance, the development of agents targeting the HER2 pathway, EGFR, VEGF, and the PI3K/Akt/mTOR signaling pathway has been particularly promising in breast cancer therapy [18]. Furthermore, the Notch and Wnt signaling pathways have been highlighted as critical targets, with ongoing studies evaluating potential inhibitors for cancer treatment [19].
In addition to traditional molecular targets, there has been a notable shift towards utilizing novel agents that modulate cancer-related signaling pathways. For example, recent reviews have emphasized the importance of targeting the Hippo signaling pathway, which is frequently dysregulated in various cancers. This pathway plays a significant role in tumorigenesis and represents a compelling target for therapeutic intervention [20].
Emerging research also underscores the relevance of immunotherapy, particularly through the use of immune checkpoint inhibitors. These therapies harness the immune system's ability to identify and eliminate cancer cells, thereby enhancing patient outcomes [21]. The integration of immunotherapy with traditional therapies has shown potential for synergistic effects, particularly in lung cancer [22].
Moreover, the exploration of epigenetic therapies and RNA-based interventions has opened new avenues for treatment. Epigenetic modulators, such as histone deacetylase (HDAC) inhibitors and DNA methyltransferase (DNMT) inhibitors, have shown promise in altering gene expression and disrupting cancer-specific signaling pathways [21]. RNA interference and mRNA-based therapies are also being investigated for their ability to target and modulate specific pathways implicated in cancer [21].
As the landscape of cancer treatment continues to evolve, it is evident that a combination of targeted therapies, immunotherapy, and novel molecular agents will be integral in developing effective treatment strategies. This multifaceted approach aims to enhance the precision of cancer therapies, reduce resistance, and improve overall patient outcomes [23].
In conclusion, the latest drug targets in cancer therapy focus on a diverse array of signaling pathways, including traditional targets like HER2 and EGFR, as well as emerging pathways such as Hippo and Notch. The ongoing research and clinical trials in these areas are expected to yield significant advancements in the fight against cancer, providing hope for improved therapeutic options.
3 Recent Advances in Targeted Therapies
3.1 Small Molecule Inhibitors
Recent advancements in targeted cancer therapies, particularly involving small molecule inhibitors (SMIs), have focused on a variety of critical molecular targets associated with tumor growth and progression. These developments reflect a growing understanding of the underlying molecular mechanisms driving cancer, leading to the identification of new therapeutic targets.
One significant area of focus is the mechanistic target of rapamycin (mTOR) signaling pathway. mTOR complexes (mTORC1 and mTORC2) regulate key cellular processes such as growth, proliferation, angiogenesis, and metabolism, which are often dysregulated in various tumors. Small molecule inhibitors targeting mTOR are currently in clinical use and in various stages of development, highlighting their importance as therapeutic agents in cancer treatment (Roohi and Hojjat-Farsangi, 2017) [24].
Another critical target includes receptor tyrosine kinases (RTKs), which play a vital role in cancer cell signaling. The development of small molecule inhibitors that specifically target these kinases has gained traction, particularly with the approval of several tyrosine kinase inhibitors (TKIs) that have shown promise in treating various malignancies. The ongoing research aims to enhance the specificity and reduce the side effects of these inhibitors, focusing on developing selective RTK-TKIs that minimize toxicity compared to multi-targeted agents (Hojjat-Farsangi, 2014) [25].
In addition, the exploration of small molecule inhibitors that target oncogenic pathways such as KRAS and cyclin-dependent kinases (CDKs) has emerged as a promising strategy. These inhibitors are designed to block the signaling pathways that are frequently altered in cancers like non-small cell lung cancer and colorectal cancer, offering potential new treatment options for patients who have limited responses to existing therapies (Kargbo, 2025) [26].
Moreover, advancements in immunotherapy have led to the development of small molecules that modulate immune responses against tumors. These agents target pathways involved in immune checkpoint regulation and other immune-related signaling pathways, representing a new frontier in cancer treatment aimed at enhancing the body's immune response to cancer cells (Wang et al., 2024) [27].
Furthermore, combination therapies that incorporate small molecule inhibitors with other treatment modalities are being actively investigated. This approach aims to address challenges such as tumor heterogeneity and resistance mechanisms, enhancing overall treatment efficacy (Gatzka, 2018) [28].
In summary, the latest drug targets for cancer treatment focus on key signaling pathways and molecular alterations associated with tumorigenesis, including mTOR, RTKs, oncogenic mutations, and immune modulation. The continuous development of small molecule inhibitors targeting these pathways represents a significant advancement in precision medicine, with the potential to improve patient outcomes through more effective and personalized cancer therapies.
3.2 Monoclonal Antibodies
Recent advancements in targeted therapies for cancer, particularly through the use of monoclonal antibodies (mAbs), have highlighted several new drug targets that are being explored in clinical settings. Monoclonal antibodies have emerged as a significant class of therapeutic agents, particularly in oncology, due to their ability to selectively target specific antigens on tumor cells and enhance the host's immune response against malignancies.
One of the primary focuses of current research involves receptor tyrosine kinases (RTKs), which play a crucial role in cancer cell signaling. Aberrant signaling through these receptors is commonly observed in various malignancies, making them attractive targets for monoclonal antibody therapy. Notably, the epidermal growth factor receptor (EGFR) and HER2 have been among the first RTKs targeted by antibody therapies, with five marketed antibodies currently available for clinical use. However, challenges such as systemic side effects, drug resistance, and refractory patient responses have prompted the exploration of new therapeutic strategies and additional RTK targets [29].
In addition to established targets like EGFR, the development of innovative monoclonal antibodies is expanding to include less well-validated antigens. This shift aims to diversify treatment options and improve therapeutic efficacy. For instance, antibody-drug conjugates (ADCs), bispecific antibodies, and engineered antibody fragments are being investigated for their potential to enhance anti-tumor responses by recruiting cytotoxic T cells or generating immune responses against cancer [30].
Furthermore, recent reviews emphasize the role of monoclonal antibodies in promoting antitumor immune responses, which is a critical area of development. Strategies that target immune cells, rather than just tumor antigens, are gaining traction as they may lead to improved outcomes in cancer therapy [31]. This innovative approach not only targets the cancer cells directly but also aims to harness the body's immune system to fight cancer more effectively.
The landscape of monoclonal antibody therapies continues to evolve, with ongoing clinical trials investigating new agents and combinations that target various cancer types. The approval of new monoclonal antibodies for conditions such as multiple myeloma exemplifies the rapid advancements in this field. In 2015 alone, four new drugs were approved, indicating a robust pipeline of mAb therapies [32].
In summary, the latest drug targets in cancer therapy, particularly through monoclonal antibodies, include established receptors like EGFR and HER2, as well as emerging targets on less well-characterized antigens. The development of novel therapeutic strategies, such as ADCs and immune-modulating antibodies, represents a significant shift in the approach to cancer treatment, with the potential to enhance efficacy and overcome resistance challenges [33].
3.3 CAR-T Cell Therapy
Recent advancements in cancer treatment have significantly focused on the development of targeted therapies, particularly through the use of chimeric antigen receptor (CAR) T cell therapy. This innovative approach utilizes genetically modified T cells to target specific cancer antigens, providing a promising strategy for both hematological malignancies and solid tumors.
Recent literature identifies a variety of novel target antigens for CAR-T cell therapy. In a review by Cao et al. (2023), several newly recognized targets for CAR-T therapy in colorectal cancer (CRC) and breast cancer (BC) were highlighted. These include GUCY2C, CLEC14A, CD26, TEM8/ANTXR1, PDPN, PTK7, PODXL, CD44, CD19, CD20, CD22, BCMA, GD2, mesothelin, TAG-72, CEA, EGFR, B7H3, HER2, IL13Ra2, MUC1, EpCAM, PSMA, and PSCA [34]. This extensive list illustrates the expanding repertoire of antigens that are being explored to enhance the efficacy of CAR-T therapies in solid tumors.
The identification of these targets is critical, especially considering the challenges faced by CAR-T therapies in treating solid tumors, such as the immunosuppressive tumor microenvironment and the heterogeneity of tumor-associated antigens [35]. As noted by Zhu (2024), CAR-T therapy has traditionally shown success in hematological cancers but is now being adapted for solid tumors, with ongoing clinical trials aimed at evaluating the efficacy of these novel targets [36].
Additionally, armored CAR-T cells, which are a fourth-generation of CAR-T therapy, have been developed to navigate the tumor microenvironment more effectively. These engineered cells can target various components of the tumor microenvironment, potentially improving the persistence and efficacy of CAR-T therapies against solid tumors [37]. This advancement represents a significant step toward overcoming the limitations of earlier CAR-T generations, which struggled with inadequate infiltration and immunosuppression.
Furthermore, innovative technologies such as next-generation sequencing are facilitating the discovery of new immune targets and mutation-derived antigens (neoantigens) for cancer immunotherapy [38]. This capability not only broadens the scope of potential therapeutic targets but also enhances the precision of immunotherapies, thereby increasing their applicability across diverse cancer types.
In summary, the landscape of drug targets for cancer therapy is rapidly evolving, particularly with CAR-T cell therapy. The identification of new antigens and the development of advanced CAR-T cell modifications are paving the way for more effective treatments, particularly in challenging solid tumors, and hold promise for improving patient outcomes in the near future.
4 Clinical Trials and Emerging Drug Targets
4.1 Overview of Current Clinical Trials
Recent advancements in cancer therapy have led to the identification of numerous novel drug targets, which are being actively investigated in clinical trials. Targeted therapies have gained prominence due to their ability to specifically interfere with molecular pathways that drive tumor growth and progression, contrasting with traditional cytotoxic treatments that affect both cancerous and normal cells.
In the realm of breast cancer, the exploration of molecularly targeted drugs has significantly expanded. The approval of agents such as lapatinib, which targets the human epidermal growth factor receptor 2 (HER2), and bevacizumab, which inhibits vascular endothelial growth factor (VEGF), marks a pivotal shift in treatment strategies. Current clinical trials are evaluating a wide range of other targeted agents, including inhibitors of the epidermal growth factor receptor (EGFR) and dual EGFR and HER2 inhibitors, as well as those targeting crucial signaling pathways such as RAS/MEK/ERK and phosphatidylinositol-3-kinase/Akt/mammalian target of rapamycin (mTOR) pathways [39].
In gynecologic cancers, the rapid development of high-throughput techniques has facilitated the identification of specific molecular targets. These include tyrosine kinases (e.g., EGFR and HER2/Neu), mTOR, and Raf kinase, which are currently under clinical investigation. Despite some setbacks, such as limited success in trials involving p53 gene therapies, the focus on molecular biology has paved the way for promising therapeutic developments [4].
Furthermore, in the broader context of oncology, the understanding of cancer biology has led to the exploration of innovative drug modalities, including nucleic acid drugs like antisense oligonucleotides (ASOs) and small interfering RNAs (siRNAs). These drugs represent a new class of therapies that can directly induce the loss of function of target genes, which is a significant shift from traditional approaches that primarily inhibit protein function [5].
Emerging drug targets also encompass a variety of signaling pathways that are critical for tumor maintenance. These include targets involved in cell invasion, apoptosis, and angiogenesis. As new molecular targets are continuously identified, the landscape of cancer therapy is rapidly evolving, with many agents currently undergoing evaluation in clinical trials. The integration of these targeted therapies with conventional treatment regimens, such as chemotherapy, is also being explored to enhance therapeutic efficacy and patient outcomes [2].
In summary, the latest drug targets in cancer therapy encompass a diverse array of molecular pathways and mechanisms. The ongoing clinical trials are crucial for validating these targets and determining their efficacy in various cancer types, thereby contributing to the advancement of personalized medicine in oncology.
4.2 Promising New Targets in Development
Recent advancements in cancer treatment have highlighted several promising new drug targets, particularly within the realm of molecularly targeted therapies. These therapies aim to disrupt specific molecular pathways that are crucial for tumor growth and progression, thereby offering a more tailored approach compared to traditional cytotoxic treatments.
One of the key focuses in recent years has been on receptor tyrosine kinases (RTKs), which are pivotal in cancer signaling pathways. Various compounds that inhibit these targets are currently under preclinical and clinical development. These include both monoclonal antibodies and small-molecule inhibitors targeting important receptors such as the epidermal growth factor receptor (EGFR), Bcr-Abl tyrosine kinase, vascular endothelial growth factor (VEGF), fibroblast growth factor receptor (FGFR), and platelet-derived growth factor (PDGF) [1]. Notably, drugs like trastuzumab and imatinib mesylate have already received FDA approval for specific indications, illustrating the potential impact of these targeted therapies on cancer care [1].
In the context of breast cancer, the landscape has evolved significantly post-trastuzumab, with new targeted therapies emerging. These include bevacizumab, which targets angiogenesis, and lapatinib, which inhibits both HER-1 and HER-2 receptors. Ongoing phase II and III clinical trials are exploring these agents, alongside other small-molecule tyrosine kinase inhibitors and mTOR inhibitors, to optimize treatment regimens [2].
Moreover, the identification of molecular targets has expanded to include not only traditional kinases but also various other pathways involved in tumor biology. The development of agents targeting specific gene expressions and signaling pathways is gaining momentum, as evidenced by the ongoing research into drugs that affect apoptosis, mitosis, and the tumor microenvironment [4]. The role of reactive oxygen species (ROS) as both markers and therapeutic targets in hepatocellular carcinoma (HCC) exemplifies this trend, with novel drug delivery systems and chemical conjugation strategies being investigated to enhance treatment efficacy [40].
Furthermore, there is an increasing interest in exploring the potential of nucleic acid drugs, such as antisense oligonucleotides (ASOs) and small interfering RNAs (siRNAs), which can directly induce the loss of function of target genes implicated in cancer [5]. This innovative approach represents a shift towards therapies that can more precisely target the molecular underpinnings of cancer.
Overall, the landscape of cancer treatment is rapidly evolving, with a focus on identifying and validating new molecular targets that can lead to the development of effective, less toxic therapies. The continuous exploration of these new avenues is critical for advancing cancer care and improving patient outcomes.
5 Challenges and Future Directions
5.1 Resistance Mechanisms
The landscape of cancer treatment is rapidly evolving, with significant advancements in the identification of novel drug targets and the development of innovative therapeutic strategies. Recent studies have highlighted various resistance mechanisms that challenge the efficacy of current treatments, necessitating a deeper understanding of these pathways to improve patient outcomes.
Recent research emphasizes the importance of targeting specific molecular alterations in cancer cells. Novel targets include cell cycle checkpoint molecules, breast cancer stem cell-related molecules, and anti-apoptotic proteins, particularly in breast cancer treatment, where drug resistance has emerged as a significant clinical challenge [41]. Additionally, mechanisms of resistance in castration-resistant prostate cancer (CRPC) have been identified, including mutations and alterations in drug targets, which necessitate the development of adjunctive therapies and new targetable pathways [42].
Emerging therapeutic strategies also involve the exploration of genetic mutations, efflux pumps, altered signaling pathways, and microenvironmental influences that contribute to drug resistance [43]. For instance, the activation of the mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K)/Akt pathways are common mechanisms through which various malignancies develop resistance to therapies [44]. This underscores the need for multi-targeted approaches and combination therapies that can address these resistance pathways more effectively.
The integration of advanced methodologies such as CRISPR-Cas9 screening has facilitated the identification of potential genetic alterations that could serve as new druggable targets [45]. This technology allows for a more nuanced understanding of the molecular complexity underpinning resistance mechanisms, thus paving the way for the design of next-generation anti-cancer drugs that can circumvent these challenges [44].
In addition to traditional small-molecule inhibitors, novel approaches utilizing nanotechnology are being explored to enhance drug delivery and efficacy while overcoming resistance [46]. The combination of targeted therapies with nanotechnology approaches presents a promising strategy to mitigate drug resistance, potentially leading to better therapeutic outcomes.
Furthermore, the recent emphasis on precision medicine and biomarker-driven strategies aims to optimize treatment responses and improve patient outcomes. By identifying specific biomarkers associated with resistance mechanisms, clinicians can tailor therapies to individual patients, thereby enhancing the likelihood of successful treatment [43].
Overall, the challenges posed by resistance mechanisms in cancer treatment underscore the need for continuous research into novel drug targets and innovative therapeutic strategies. The evolving understanding of cancer biology, combined with technological advancements, offers hope for the development of more effective treatments that can overcome the hurdles of drug resistance.
5.2 Personalized Medicine Approaches
The latest advancements in cancer therapy have increasingly focused on personalized medicine approaches, which aim to tailor treatments based on the unique genetic and molecular profiles of individual tumors. Recent studies highlight several novel drug targets and the evolving landscape of targeted therapies, alongside the challenges and future directions in this field.
Recent insights from cancer genomics have unveiled novel points of attack for targeted cancer therapy. The Cancer Genome Atlas and the International Cancer Genome Consortium have provided comprehensive genomic characterizations, revealing that individual cancers often contain only a few recurrent driver genes. This knowledge has accelerated the identification of new drug targets, facilitating the development of specific molecular therapies. The use of cost-effective sequencing technologies enables comprehensive mutational profiling, which is crucial for the revolution of personalized cancer medicine in the coming years (Røsland & Engelsen, 2015) [47].
In the realm of personalized cancer medicine, molecularly distinct subtypes of cancers have been identified, necessitating different therapeutic strategies for each subtype. For instance, targeted therapies such as monoclonal antibodies (trastuzumab for HER2-positive breast cancer) and tyrosine kinase inhibitors (imatinib for chronic myeloid leukemia) exemplify how treatment decisions are increasingly based on the molecular abnormality profile of tumors rather than solely on tissue type (Jackson & Chester, 2015) [48].
Moreover, the introduction of targeted therapies has marked a significant shift towards personalized treatment in older patients with solid tumors, as understanding tumor heterogeneity allows for the identification of specific targets for treatment (Kelly et al., 2014) [49]. Despite these advancements, the implementation of personalized medicine faces substantial challenges, including the high cost of targeted therapies and the need for extensive molecular profiling, which can be limited by the availability of specific drugs and the complexity of tumor biology.
Future directions in personalized cancer therapy will likely involve overcoming these challenges by refining molecular profiling techniques and developing combination therapies. The I-PREDICT study exemplifies this approach, demonstrating that personalized treatment with combination therapies can improve outcomes in patients with refractory malignancies. This study showed that targeting a larger fraction of identified molecular alterations correlated with significantly improved disease control rates and overall survival, suggesting that a multi-targeted approach may be more effective than single-agent therapies (Sicklick et al., 2019) [50].
In summary, the field of personalized cancer medicine is rapidly evolving, with new drug targets emerging from advanced genomic insights. However, to fully realize the potential of these therapies, ongoing efforts are needed to address the existing challenges and optimize treatment strategies for diverse cancer patient populations.
5.3 Combination Therapies
The landscape of cancer treatment has evolved significantly, particularly with the increasing emphasis on combination therapies that utilize targeted drugs. The emergence of novel drug targets and combination strategies has been shaped by the need to address challenges such as tumor heterogeneity, drug resistance, and the limitations of monotherapy.
Recent advancements highlight the importance of multi-targeted agents in overcoming the complexities associated with cancer treatment. For instance, the use of small-molecule drugs, particularly multi-targeted kinase inhibitors, has shown promise in combination therapies. These agents can target multiple signaling pathways simultaneously, which is crucial given the intricate network of molecular interactions involved in cancer progression. This strategy aims to enhance therapeutic efficacy while reducing the likelihood of resistance (Mologni et al., 2021) [51].
Combination therapies are gaining traction as they leverage different mechanisms of action to improve treatment outcomes. For example, the integration of DNA damage repair (DDR) inhibitors with immunotherapies has been shown to significantly enhance therapeutic effects, especially in solid tumors that are typically resistant to conventional treatments (Wang et al., 2022) [52]. The rationale behind this approach is to exploit the synergistic effects of combining therapies that target distinct but complementary pathways.
Moreover, the development of hybrid compounds that incorporate multiple bioactive moieties into a single entity is emerging as a promising strategy. These hybrid agents can address multiple targets simultaneously, potentially leading to improved efficacy and reduced side effects (Kucuksayan & Ozben, 2017) [53]. The design of such multitargeted agents is particularly important in the context of cancer, where single-target therapies often fall short due to the complex genetic and environmental interactions that drive tumor growth.
In addition to hybrid compounds, fixed-dose combination (FDC) drugs are gaining attention. These formulations are designed to synergistically target multiple sites, which is particularly relevant for treating complex diseases like cancer. Ongoing research is focused on refining methods for evaluating drug synergism, which is crucial for advancing combination drug discovery (Vottero et al., 2025) [54].
The clinical application of combination therapies is supported by ongoing trials that investigate various combinations of molecular-targeted agents (MATs) with cytotoxic agents. This approach aims to inhibit multiple pathways simultaneously, thereby enhancing the overall anticancer efficacy (Li et al., 2014) [55]. The integration of immunotherapies with traditional chemotherapy is also being explored, with the goal of improving patient responses and overcoming resistance mechanisms.
Despite these advancements, challenges remain, particularly in addressing the high degree of clonal heterogeneity within tumors and the complex interplay of signaling pathways. Future directions in cancer therapy will likely focus on personalized medicine, where treatment regimens are tailored based on individual patient parameters and tumor characteristics, as well as the continued exploration of innovative combination strategies to improve outcomes for patients with refractory cancers.
In conclusion, the future of cancer therapy lies in the continued development of targeted combination therapies that address the multifaceted nature of cancer. This approach not only aims to improve therapeutic efficacy but also seeks to minimize the adverse effects associated with traditional treatments, ultimately leading to better patient outcomes.
6 Conclusion
The exploration of novel drug targets in cancer therapy has unveiled significant advancements that promise to reshape treatment paradigms. Key findings indicate that oncogenes, tumor suppressor genes, immune checkpoints, and novel signaling pathways represent critical areas for targeted interventions. The integration of small molecule inhibitors, monoclonal antibodies, and CAR-T cell therapies exemplifies the shift towards more precise cancer treatments. However, challenges such as drug resistance and tumor heterogeneity continue to complicate therapeutic efficacy. Future research should focus on overcoming these obstacles through combination therapies and personalized medicine approaches, which aim to tailor treatments based on individual tumor profiles. By leveraging advanced genomic insights and innovative drug delivery systems, the potential for improved patient outcomes in cancer therapy remains promising.
References
- [1] Sanjay Goel;Sridhar Mani;Roman Perez-Soler. Tyrosine kinase inhibitors: a clinical perspective.. Current oncology reports(IF=5.0). 2002. PMID:11734109. DOI: 10.1007/s11912-002-0043-x.
- [2] Fausto Petrelli;Mary Cabiddu;Marina Elena Cazzaniga;Marco Cremonesi;Sandro Barni. Targeted therapies for the treatment of breast cancer in the post-trastuzumab era.. The oncologist(IF=4.2). 2008. PMID:18448551. DOI: 10.1634/theoncologist.2007-0173.
- [3] P Workman. Scoring a bull's-eye against cancer genome targets.. Current opinion in pharmacology(IF=4.2). 2001. PMID:11710731. DOI: 10.1016/s1471-4892(01)00060-1.
- [4] Hye Sook Chon;Wei Hu;John J Kavanagh. Targeted therapies in gynecologic cancers.. Current cancer drug targets(IF=3.5). 2006. PMID:16848724. DOI: 10.2174/156800906777441799.
- [5] Shota Oyama;Tsuyoshi Yamamoto;Asako Yamayoshi. Recent Advances in the Delivery Carriers and Chemical Conjugation Strategies for Nucleic Acid Drugs.. Cancers(IF=4.4). 2021. PMID:34359781. DOI: 10.3390/cancers13153881.
- [6] Ryuya Yamanaka;Hideyuki Saya. Molecularly targeted therapies for glioma.. Annals of neurology(IF=7.7). 2009. PMID:20035507. DOI: 10.1002/ana.21793.
- [7] Daniela D Rosa;Gustavo Ismael;Lissandra Dal Lago;Ahmad Awada. Molecular-targeted therapies: lessons from years of clinical development.. Cancer treatment reviews(IF=10.5). 2008. PMID:17826917. DOI: 10.1016/j.ctrv.2007.07.019.
- [8] Edgar Selzer;Gabriela Kornek. Targeted drugs in combination with radiotherapy for the treatment of solid tumors: current state and future developments.. Expert review of clinical pharmacology(IF=3.0). 2013. PMID:24164614. DOI: 10.1586/17512433.2013.841540.
- [9] L Xu;K F Pirollo;E H Chang. Tumor-targeted p53-gene therapy enhances the efficacy of conventional chemo/radiotherapy.. Journal of controlled release : official journal of the Controlled Release Society(IF=11.5). 2001. PMID:11489488. DOI: 10.1016/s0168-3659(01)00324-8.
- [10] Jia Yee Lee;Richie R Bhandare;Sai H S Boddu;Afzal B Shaik;Lakshmana Prabu Saktivel;Gaurav Gupta;Poonam Negi;Muna Barakat;Sachin Kumar Singh;Kamal Dua;Dinesh Kumar Chellappan. Molecular mechanisms underlying the regulation of tumour suppressor genes in lung cancer.. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie(IF=7.5). 2024. PMID:38394846. DOI: 10.1016/j.biopha.2024.116275.
- [11] Ezharul Hoque Chowdhury. Strategies for tumor-directed delivery of siRNA.. Expert opinion on drug delivery(IF=5.4). 2011. PMID:21314230. DOI: 10.1517/17425247.2011.554817.
- [12] Dulcie Lai;Stacy Visser-Grieve;Xiaolong Yang. Tumour suppressor genes in chemotherapeutic drug response.. Bioscience reports(IF=4.7). 2012. PMID:22762204. DOI: 10.1042/BSR20110125.
- [13] Robert J Torphy;Richard D Schulick;Yuwen Zhu. Newly Emerging Immune Checkpoints: Promises for Future Cancer Therapy.. International journal of molecular sciences(IF=4.9). 2017. PMID:29211042. DOI: 10.3390/ijms18122642.
- [14] Leticia De Mattos-Arruda;Juan Blanco-Heredia;Carmen Aguilar-Gurrieri;Jorge Carrillo;Julià Blanco. New emerging targets in cancer immunotherapy: the role of neoantigens.. ESMO open(IF=8.3). 2020. PMID:32269031. DOI: 10.1136/esmoopen-2020-000684.
- [15] Zlatko Dembic. Antitumor Drugs and Their Targets.. Molecules (Basel, Switzerland)(IF=4.6). 2020. PMID:33297561. DOI: 10.3390/molecules25235776.
- [16] Lingjiao Meng;Haotian Wu;Jiaxiang Wu;Ping'an Ding;Jinchen He;Meixiang Sang;Lihua Liu. Mechanisms of immune checkpoint inhibitors: insights into the regulation of circular RNAS involved in cancer hallmarks.. Cell death & disease(IF=9.6). 2024. PMID:38177102. DOI: 10.1038/s41419-023-06389-5.
- [17] Hamidreza Akhbariyoon;Yasaman Azizpour;Marjan Fakhrizadeh Esfahani;Maryam Sadat Mirbagheri Firoozabad;Mehrdad Rabiee Rad;Kiarash Sadeghian Esfahani;Neda Khoshavi;Negin Karimi;Asal Shirinisaz;Fatemeh Abedi;Maryam Rabiee Rad;Parisa Sharifi. Immune checkpoint inhibition for the treatment of cancers: An update and critical review of ongoing clinical trials.. Clinical immunology (Orlando, Fla.)(IF=3.8). 2021. PMID:34688855. DOI: 10.1016/j.clim.2021.108873.
- [18] Máximo de la Vega;Enrique Díaz-Cantón;Ricardo H Alvarez. Novel targeted agents for the treatment of advanced breast cancer.. Future medicinal chemistry(IF=3.4). 2012. PMID:22571614. DOI: 10.4155/fmc.12.45.
- [19] Yu-Feng Xiao;Xin Yong;Bo Tang;Yong Qin;Jian-Wei Zhang;Dan Zhang;Rui Xie;Shi-Ming Yang. Notch and Wnt signaling pathway in cancer: Crucial role and potential therapeutic targets (Review).. International journal of oncology(IF=4.9). 2016. PMID:26648421. DOI: 10.3892/ijo.2015.3280.
- [20] Rui Zhu;Zhihan Jiao;Fa-Xing Yu. Advances towards potential cancer therapeutics targeting Hippo signaling.. Biochemical Society transactions(IF=4.3). 2024. PMID:39641583. DOI: 10.1042/BST20240244.
- [21] A S Shifana;Mohammad Adnan;Akash Gupta; Ajazuddin;Parag Jain. A Comprehensive Review on Novel Pathways in Cancer Treatment: Clinical Applications and Future Prospects.. Current cancer drug targets(IF=3.5). 2025. PMID:39069707. DOI: 10.2174/0115680096312603240709112520.
- [22] William C S Cho. Targeting signaling pathways in lung cancer therapy.. Expert opinion on therapeutic targets(IF=4.4). 2013. PMID:23231077. DOI: 10.1517/14728222.2013.729043.
- [23] Hon Yan Kelvin Yip;Antonella Papa. Signaling Pathways in Cancer: Therapeutic Targets, Combinatorial Treatments, and New Developments.. Cells(IF=5.2). 2021. PMID:33809714. DOI: 10.3390/cells10030659.
- [24] Azam Roohi;Mohammad Hojjat-Farsangi. Recent advances in targeting mTOR signaling pathway using small molecule inhibitors.. Journal of drug targeting(IF=3.9). 2017. PMID:27632356. DOI: 10.1080/1061186X.2016.1236112.
- [25] Mohammad Hojjat-Farsangi. Small-molecule inhibitors of the receptor tyrosine kinases: promising tools for targeted cancer therapies.. International journal of molecular sciences(IF=4.9). 2014. PMID:25110867. DOI: 10.3390/ijms150813768.
- [26] Robert B Kargbo. Targeting Oncogenic Pathways: Advances in KRAS, CDK, EGFR, and PROTAC-Based Therapies.. ACS medicinal chemistry letters(IF=4.0). 2025. PMID:40236546. DOI: 10.1021/acsmedchemlett.5c00111.
- [27] Fang Wang;Kai Fu;Yujue Wang;Can Pan;Xueping Wang;Zeyu Liu;Chuan Yang;Ying Zheng;Xiaopeng Li;Yu Lu;Kenneth Kin Wah To;Chenglai Xia;Jianye Zhang;Zhi Shi;Zeping Hu;Min Huang;Liwu Fu. Small-molecule agents for cancer immunotherapy.. Acta pharmaceutica Sinica. B(IF=14.6). 2024. PMID:38486980. DOI: 10.1016/j.apsb.2023.12.010.
- [28] Martina V Gatzka. Targeted Tumor Therapy Remixed-An Update on the Use of Small-Molecule Drugs in Combination Therapies.. Cancers(IF=4.4). 2018. PMID:29794999. DOI: 10.3390/cancers10060155.
- [29] Bénédicte Fauvel;Aziz Yasri. Antibodies directed against receptor tyrosine kinases: current and future strategies to fight cancer.. mAbs(IF=7.3). 2014. PMID:24859229. DOI: 10.4161/mabs.29089.
- [30] Janice M Reichert;Eugen Dhimolea. The future of antibodies as cancer drugs.. Drug discovery today(IF=7.5). 2012. PMID:22561895. DOI: 10.1016/j.drudis.2012.04.006.
- [31] Louis M Weiner;Joseph C Murray;Casey W Shuptrine. Antibody-based immunotherapy of cancer.. Cell(IF=42.5). 2012. PMID:22424219. DOI: 10.1016/j.cell.2012.02.034.
- [32] Elizabeth K O'Donnell;Noopur S Raje. New monoclonal antibodies on the horizon in multiple myeloma.. Therapeutic advances in hematology(IF=3.1). 2017. PMID:28203341. DOI: 10.1177/2040620716682490.
- [33] Divaya Bhutani;Ulka N Vaishampayan. Monoclonal antibodies in oncology therapeutics: present and future indications.. Expert opinion on biological therapy(IF=4.0). 2013. PMID:23286740. DOI: 10.1517/14712598.2012.758705.
- [34] Yu Cao;Sergey K Efetov;Mingze He;Yu Fu;Narasimha M Beeraka;Jin Zhang;Xinliang Zhang;Namitha Bannimath;Kuo Chen. Updated Clinical Perspectives and Challenges of Chimeric Antigen Receptor-T Cell Therapy in Colorectal Cancer and Invasive Breast Cancer.. Archivum immunologiae et therapiae experimentalis(IF=3.9). 2023. PMID:37566162. DOI: 10.1007/s00005-023-00684-x.
- [35] Kai Cui;Wanjun He;Na Huang;Songshan Zhu;Dan Jiang;Weiqiang Yang;Yiwei Zeng;Muhammad Asad Farooq;Guangxian Xu. The challenges and progress of CAR-T cell therapy in the treatment of solid tumors.. Molecular and cellular biochemistry(IF=3.7). 2025. PMID:40549304. DOI: 10.1007/s11010-025-05329-5.
- [36] Shigui Zhu. CAR-T in cancer therapeutics and updates.. Journal of the National Cancer Center(IF=9.4). 2024. PMID:39281717. DOI: 10.1016/j.jncc.2024.01.001.
- [37] Xianjun Li;Tianjun Chen;Xuehan Li;Hanyu Zhang;Yingjing Li;Shuyuan Zhang;Shengnan Luo;Tongsen Zheng. Therapeutic targets of armored chimeric antigen receptor T cells navigating the tumor microenvironment.. Experimental hematology & oncology(IF=13.5). 2024. PMID:39350256. DOI: 10.1186/s40164-024-00564-w.
- [38] Rong-Fu Wang;Helen Y Wang. Immune targets and neoantigens for cancer immunotherapy and precision medicine.. Cell research(IF=25.9). 2017. PMID:28025978. DOI: 10.1038/cr.2016.155.
- [39] Ricardo H Alvarez;Vicente Valero;Gabriel N Hortobagyi. Emerging targeted therapies for breast cancer.. Journal of clinical oncology : official journal of the American Society of Clinical Oncology(IF=41.9). 2010. PMID:20530283. DOI: 10.1200/JCO.2009.25.4011.
- [40] Lin Xing;Yuting Tang;Lu Li;Xufeng Tao. ROS in hepatocellular carcinoma: What we know.. Archives of biochemistry and biophysics(IF=3.0). 2023. PMID:37499994. DOI: 10.1016/j.abb.2023.109699.
- [41] Ruixian Chen;Zhilan Zhou;Xiangyue Meng;Yi Lei;Yuxi Wang;Yanyan Wang. Emerging opportunities to treat drug-resistant breast cancer: Discovery of novel small-molecule inhibitors against different targets.. Frontiers in pharmacology(IF=4.8). 2025. PMID:40949156. DOI: 10.3389/fphar.2025.1578342.
- [42] Thenappan Chandrasekar;Joy C Yang;Allen C Gao;Christopher P Evans. Targeting molecular resistance in castration-resistant prostate cancer.. BMC medicine(IF=8.3). 2015. PMID:26329698. DOI: 10.1186/s12916-015-0457-6.
- [43] Pankaj Garg;Jyoti Malhotra;Prakash Kulkarni;David Horne;Ravi Salgia;Sharad S Singhal. Emerging Therapeutic Strategies to Overcome Drug Resistance in Cancer Cells.. Cancers(IF=4.4). 2024. PMID:39001539. DOI: 10.3390/cancers16132478.
- [44] 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.
- [45] Jue Hou;Zongsheng He;Tian Liu;Dongfeng Chen;Bin Wang;Qinglian Wen;Xi Zheng. Evolution of Molecular Targeted Cancer Therapy: Mechanisms of Drug Resistance and Novel Opportunities Identified by CRISPR-Cas9 Screening.. Frontiers in oncology(IF=3.3). 2022. PMID:35372044. DOI: 10.3389/fonc.2022.755053.
- [46] 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.
- [47] Gro Vatne Røsland;Agnete Svendsen Tenfjord Engelsen. Novel points of attack for targeted cancer therapy.. Basic & clinical pharmacology & toxicology(IF=3.3). 2015. PMID:25154903. DOI: 10.1111/bcpt.12313.
- [48] Sarah E Jackson;John D Chester. Personalised cancer medicine.. International journal of cancer(IF=4.7). 2015. PMID:24789362. DOI: 10.1002/ijc.28940.
- [49] Ciara M Kelly;Derek G Power;Stuart M Lichtman. Targeted therapy in older patients with solid tumors.. Journal of clinical oncology : official journal of the American Society of Clinical Oncology(IF=41.9). 2014. PMID:25071113. DOI: 10.1200/JCO.2014.55.4246.
- [50] 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.
- [51] Luca Mologni;Giovanni Marzaro;Sara Redaelli;Alfonso Zambon. Dual Kinase Targeting in Leukemia.. Cancers(IF=4.4). 2021. PMID:33401428. DOI: 10.3390/cancers13010119.
- [52] Yumin Wang;Meihan Duan;Zhouying Peng;Ruohao Fan;Yuxiang He;Hua Zhang;Wei Xiong;Weihong Jiang. Advances of DNA Damage Repair-Related Drugs and Combination With Immunotherapy in Tumor Treatment.. Frontiers in immunology(IF=5.9). 2022. PMID:35281059. DOI: 10.3389/fimmu.2022.854730.
- [53] Ertan Kucuksayan;Tomris Ozben. Hybrid Compounds as Multitarget Directed Anticancer Agents.. Current topics in medicinal chemistry(IF=3.3). 2017. PMID:27697050. DOI: 10.2174/1568026616666160927155515.
- [54] Paola Vottero;Jack Tuszynski;Yun K Tam;Chih-Yuan Tseng. Synergism or mirage: Current progress and an empirical approach for elucidating combination drug effects.. Drug discovery today(IF=7.5). 2025. PMID:40383289. DOI: 10.1016/j.drudis.2025.104381.
- [55] Feifei Li;Changqi Zhao;Lili Wang. Molecular-targeted agents combination therapy for cancer: developments and potentials.. International journal of cancer(IF=4.7). 2014. PMID:23649791. DOI: 10.1002/ijc.28261.
MaltSci Intelligent Research Services
Search for more papers on MaltSci.com
cancer drug targets · targeted therapies · personalized medicine
© 2025 MaltSci
