Skip to content

This report is written by MaltSci based on the latest literature and research findings


What are the mechanisms of cancer drug resistance?

Abstract

Cancer remains a leading cause of morbidity and mortality globally, and drug resistance poses a significant barrier to effective treatment. Understanding the mechanisms underlying cancer drug resistance is crucial for enhancing therapeutic strategies and improving patient outcomes. This review explores the multifaceted biological and molecular mechanisms contributing to drug resistance, including genetic mutations, epigenetic modifications, interactions within the tumor microenvironment, and the role of cancer stem cells. Genetic mechanisms of resistance primarily involve mutations in drug targets, leading to altered drug binding and activation of compensatory pathways that allow tumor cells to survive despite treatment. Epigenetic changes, such as DNA methylation and histone modifications, further complicate resistance by affecting gene expression patterns critical for drug metabolism and apoptosis. The tumor microenvironment significantly influences drug response, with stromal cells and hypoxic conditions promoting survival and resistance through various signaling pathways. Additionally, cancer stem cells exhibit unique characteristics that enhance their ability to withstand therapies, including self-renewal, enhanced DNA repair, and expression of drug efflux pumps. Clinical implications of these findings suggest the need for predictive biomarkers to tailor treatment strategies and the potential of combination therapies to target multiple resistance mechanisms simultaneously. Ultimately, a comprehensive understanding of the intricate mechanisms of drug resistance will pave the way for innovative therapeutic approaches that improve efficacy and patient survival in the ongoing battle against cancer.

Outline

This report will discuss the following questions.

  • 1 Introduction
  • 2 Genetic Mechanisms of Drug Resistance
    • 2.1 Mutations in Drug Targets
    • 2.2 Activation of Alternative Pathways
  • 3 Epigenetic Modifications
    • 3.1 DNA Methylation Changes
    • 3.2 Histone Modification Patterns
  • 4 Tumor Microenvironment and Drug Resistance
    • 4.1 Role of Stromal Cells
    • 4.2 Hypoxia and Nutrient Availability
  • 5 Cancer Stem Cells and Resistance
    • 5.1 Characteristics of Cancer Stem Cells
    • 5.2 Mechanisms of Resistance in Cancer Stem Cells
  • 6 Clinical Implications and Strategies to Overcome Resistance
    • 6.1 Predictive Biomarkers
    • 6.2 Combination Therapies
  • 7 Summary

1 Introduction

Cancer remains one of the leading causes of morbidity and mortality worldwide, presenting a formidable challenge to modern medicine. Despite advancements in therapeutic strategies, including chemotherapy and targeted therapies, the development of drug resistance significantly undermines treatment efficacy and contributes to poor patient outcomes. Understanding the intricate mechanisms underlying cancer drug resistance is crucial for improving therapeutic strategies and ultimately enhancing patient survival. This review aims to elucidate the multifaceted biological and molecular mechanisms that contribute to cancer drug resistance, including genetic mutations, epigenetic modifications, interactions within the tumor microenvironment, and the role of cancer stem cells. By providing a comprehensive overview of these mechanisms, we can identify potential therapeutic targets and strategies to overcome resistance.

The significance of addressing cancer drug resistance cannot be overstated. Resistance can arise through various pathways, including alterations in drug targets, activation of compensatory survival pathways, and changes in drug metabolism [1][2]. Furthermore, the tumor microenvironment plays a pivotal role in modulating drug response, influencing both intrinsic and extrinsic factors that dictate therapeutic efficacy [3][4]. The emergence of drug-resistant cancer cell populations often leads to treatment failure, disease progression, and ultimately, increased mortality [1][1]. Therefore, a thorough understanding of the mechanisms that drive resistance is essential for the development of innovative treatment modalities and personalized medicine approaches.

Current research has identified several key mechanisms of drug resistance, including genetic mutations that affect drug targets, alterations in drug transport and metabolism, and epigenetic modifications that regulate gene expression [5][6]. Additionally, tumor heterogeneity and the presence of cancer stem cells contribute to the complexity of resistance, as these cells can possess unique properties that enable them to survive conventional therapies [3][4]. The interplay between these factors creates a dynamic landscape of resistance that complicates treatment strategies and necessitates ongoing investigation.

This review is organized as follows: we will first explore the genetic mechanisms of drug resistance, detailing mutations in drug targets and the activation of alternative signaling pathways. Next, we will examine the role of epigenetic modifications, focusing on DNA methylation changes and histone modification patterns that influence gene expression. Following this, we will discuss the tumor microenvironment's impact on drug resistance, highlighting the roles of stromal cells and factors such as hypoxia and nutrient availability. The characteristics and mechanisms of resistance associated with cancer stem cells will be addressed next, followed by a discussion of clinical implications and strategies to overcome resistance, including the identification of predictive biomarkers and the use of combination therapies. Finally, we will summarize the key findings and suggest future directions for research in this critical area.

In conclusion, unraveling the mechanisms of cancer drug resistance is paramount for advancing cancer therapy. By gaining insights into the complex interplay of genetic, epigenetic, and microenvironmental factors, we can develop more effective treatment strategies that not only target the tumor itself but also address the underlying mechanisms that contribute to resistance. This comprehensive understanding will pave the way for innovative approaches that enhance therapeutic efficacy and improve patient outcomes in the fight against cancer.

2 Genetic Mechanisms of Drug Resistance

2.1 Mutations in Drug Targets

Cancer drug resistance is a multifaceted phenomenon that arises from various genetic mechanisms, prominently including mutations in drug targets. These mutations can significantly alter the efficacy of therapeutic agents, leading to treatment failure.

One of the primary mechanisms of drug resistance involves secondary mutations in drug targets. These mutations can lead to changes in the binding affinity of the drug, rendering it less effective or entirely ineffective. For instance, in the context of targeted therapies, mutations may occur in oncogenes or tumor suppressor genes that are the intended targets of these drugs, ultimately resulting in resistance. Research indicates that up to 63% of somatic mutations can exhibit heterogeneity within individual tumors, which contributes to the development of resistance (Malgorzata Roszkowska, 2024) [7].

In addition to mutations in drug targets, genetic alterations can also activate alternative signaling pathways that bypass the inhibited pathway, further complicating treatment efforts. For example, tumors may develop compensatory mechanisms that allow them to survive despite the presence of targeted therapies, effectively utilizing alternate routes for growth and survival (Michael M Gottesman et al., 2016) [6].

Moreover, the tumor microenvironment plays a crucial role in the manifestation of drug resistance. Factors within the microenvironment, such as the presence of cancer-associated fibroblasts and immune cell populations, can influence the expression of genes related to drug metabolism and efflux, contributing to the overall resistance profile of the tumor (Sudikshaa Vijayakumar et al., 2024) [1].

Epigenetic changes also interact with genetic mutations to facilitate resistance. Alterations in DNA methylation and histone modifications can lead to the aberrant expression of genes involved in drug metabolism, repair mechanisms, and apoptosis, thereby promoting a drug-resistant phenotype (Roel H Wilting and Jan-Hermen Dannenberg, 2012) [8]. This epigenetic regulation can result in a dynamic and heterogeneous tumor cell population, which can adapt to therapeutic pressures, making it challenging to achieve sustained responses to treatment.

Overall, the genetic mechanisms of cancer drug resistance are complex and interconnected, involving mutations in drug targets, activation of alternative pathways, and the influence of the tumor microenvironment. These insights highlight the necessity for comprehensive strategies that target multiple resistance mechanisms simultaneously, which may improve treatment outcomes in cancer therapy.

2.2 Activation of Alternative Pathways

Cancer drug resistance is a multifaceted phenomenon that significantly impedes effective treatment outcomes. One of the critical mechanisms underlying this resistance involves the activation of alternative signaling pathways. This activation can occur due to various factors, including genetic mutations, epigenetic changes, and alterations in the tumor microenvironment.

The activation of alternative pathways often serves as a compensatory mechanism when the primary drug target is inhibited or when the cancer cells develop mutations that render them less sensitive to the treatment. For instance, cancer cells may activate compensatory survival pathways, allowing them to bypass the effects of the drug and continue proliferating. This is particularly evident in cases where targeted therapies are employed, as cancer cells can exploit alternative growth and survival pathways to maintain their viability despite the presence of the therapeutic agent.

In addition to genetic mutations, the tumor microenvironment plays a crucial role in modulating these pathways. Factors within the microenvironment, such as cytokines and growth factors, can promote the activation of alternative signaling cascades that enhance cell survival and drug resistance. The interplay between the tumor cells and their microenvironment is complex, as the latter can influence tumor heterogeneity and the dynamics of drug response.

Furthermore, research has identified specific signaling pathways that are frequently involved in drug resistance. For example, pathways such as the phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) and mitogen-activated protein kinase (MAPK) pathways are often hyperactivated in resistant cancer cells, contributing to their survival and proliferation in the face of therapeutic challenges [9].

Moreover, the emergence of polytherapy approaches targeting multiple resistance mechanisms has been proposed as a strategy to overcome this challenge. By simultaneously inhibiting various pathways that contribute to resistance, it may be possible to enhance the effectiveness of cancer therapies and improve patient outcomes [3].

In summary, the activation of alternative pathways is a key mechanism of cancer drug resistance, driven by genetic alterations, epigenetic modifications, and the influence of the tumor microenvironment. Understanding these mechanisms is essential for developing innovative therapeutic strategies aimed at overcoming resistance and improving the efficacy of cancer treatments [1][10][11].

3 Epigenetic Modifications

3.1 DNA Methylation Changes

Cancer drug resistance is a multifaceted phenomenon that significantly complicates the treatment of malignancies. One of the key mechanisms contributing to drug resistance is the alteration of epigenetic modifications, particularly changes in DNA methylation patterns. DNA methylation, a common epigenetic mechanism, involves the addition of methyl groups to the DNA molecule, which can affect gene expression without altering the underlying nucleotide sequence.

Research indicates that aberrant DNA methylation is frequently observed in cancer cells, leading to both hypermethylation of tumor suppressor genes and hypomethylation of oncogenes. These changes can disrupt normal regulatory mechanisms, contributing to the survival and proliferation of cancer cells in the presence of therapeutic agents. For instance, hypermethylation of genes that normally act to suppress tumor growth can lead to their silencing, thereby allowing uncontrolled cell division and tumor progression [12].

Moreover, the global loss of DNA methylation can activate oncogenes, further promoting tumorigenesis and resistance to treatment [12]. The dynamic nature of these epigenetic modifications allows cancer cells to adapt to therapeutic pressures, creating a heterogeneous population of cells within a tumor. This heterogeneity can lead to the selection of drug-resistant clones, which survive initial treatment and contribute to disease recurrence [8].

In breast cancer, for example, genetic alterations, including mutations in oncogenes and tumor suppressor genes, interact with epigenetic changes to facilitate resistance to both chemotherapy and targeted therapies. The interplay between genetic and epigenetic modifications can result in a reprogramming of gene expression that supports adaptive resistance mechanisms [13]. Additionally, specific alterations in DNA methylation patterns have been identified as potential biomarkers for predicting responses to chemotherapy, suggesting that monitoring these changes could inform treatment strategies [12].

The reversibility of DNA methylation presents a unique opportunity for therapeutic intervention. Epigenetic drugs, such as DNA methyltransferase inhibitors, have been developed to target these modifications and potentially restore normal gene expression patterns in cancer cells. Such treatments aim to reverse the epigenetic silencing of tumor suppressor genes and enhance the sensitivity of cancer cells to existing therapies [14].

In summary, DNA methylation changes play a critical role in the development of cancer drug resistance. The ability of cancer cells to alter their epigenetic landscape allows them to evade therapeutic interventions, making it essential to understand these mechanisms to develop more effective treatment strategies. Future research is needed to further elucidate the specific epigenetic alterations associated with drug resistance and to explore the potential of epigenetic therapies in overcoming these challenges [15][16].

3.2 Histone Modification Patterns

Cancer drug resistance is a multifaceted challenge in oncology, with various mechanisms contributing to the ineffectiveness of therapeutic agents. Among these, epigenetic modifications, particularly histone modifications, play a critical role in mediating drug resistance in cancer cells. Histone modifications are reversible changes that affect chromatin structure and gene expression without altering the underlying DNA sequence, making them significant players in the regulation of cellular responses to therapy.

One primary mechanism by which histone modifications contribute to drug resistance is through the alteration of gene expression profiles. For instance, histone deacetylases (HDACs) are enzymes that remove acetyl groups from histones, leading to a more compact chromatin structure and reduced transcriptional activity of associated genes. This can result in the downregulation of pro-apoptotic factors and upregulation of drug transporters, enabling cancer cells to evade the cytotoxic effects of chemotherapeutics. The activity of HDACs has been linked to resistance in various cancer types, where they influence the expression of genes involved in cell survival and proliferation, thereby facilitating a resistant phenotype (Minisini et al. 2024; Kwon et al. 2018).

Additionally, histone methylation, another form of histone modification, is associated with the regulation of gene expression and has been implicated in the development of drug resistance. For example, the overexpression of histone lysine demethylases (KDMs) has been observed in several cancers, where they modify the methylation status of histones, thereby influencing the expression of genes that promote survival and resistance to therapies such as tyrosine kinase inhibitors (White et al. 2019). Specifically, KDM5A, a histone lysine demethylase, has been shown to mediate resistance to EGFR inhibitors in lung cancer by activating alternative survival pathways (White et al. 2019).

Moreover, aberrant DNA methylation patterns, often coupled with histone modifications, can lead to the silencing of tumor suppressor genes and the activation of oncogenes, further contributing to drug resistance. These epigenetic alterations create a dynamic and heterogeneous tumor microenvironment that can adapt to therapeutic pressures, resulting in the selection of drug-resistant clones (Wang et al. 2023).

The reversible nature of histone modifications presents opportunities for therapeutic intervention. Epigenetic drugs, such as HDAC inhibitors and DNA methyltransferase inhibitors, aim to restore normal epigenetic regulation and potentially reverse drug resistance. Recent studies have highlighted the efficacy of combining these agents with conventional therapies to enhance sensitivity to treatment and overcome resistance mechanisms (Quagliano et al. 2020; Alalhareth et al. 2025).

In summary, histone modifications are pivotal in the mechanisms underlying cancer drug resistance. By regulating gene expression and contributing to the dynamic epigenetic landscape of tumors, these modifications facilitate the survival of cancer cells in the presence of therapeutic agents. Understanding these mechanisms not only provides insights into the biology of drug resistance but also opens avenues for the development of novel therapeutic strategies aimed at reversing resistance and improving patient outcomes.

4 Tumor Microenvironment and Drug Resistance

4.1 Role of Stromal Cells

Cancer drug resistance is a multifaceted phenomenon significantly influenced by the tumor microenvironment (TME), particularly through the interactions involving stromal cells. The TME encompasses various components, including normal stromal cells, extracellular matrix, and soluble factors such as cytokines and growth factors, which collectively contribute to drug resistance in cancer therapy.

One of the primary mechanisms of drug resistance attributed to the TME is the concept of environment-mediated drug resistance (EM-DR). This encompasses two major forms: cell adhesion-mediated drug resistance (CAM-DR) and soluble factor-mediated drug resistance (SM-DR). CAM-DR occurs when tumor cells interact directly with stromal cells or extracellular matrix components, leading to the activation of signaling pathways that promote cell survival and inhibit drug-induced apoptosis. For instance, binding of tumor cells to integrins on stromal cells can trigger intracellular signaling cascades that enhance cell viability in the presence of chemotherapeutic agents [17].

Stromal cells, such as cancer-associated fibroblasts (CAFs), play a crucial role in this process. They can secrete soluble factors like interleukins that enhance tumor cell survival, thus contributing to the overall resistance observed in tumors. For example, interleukin-6 (IL-6) is known to enhance tumor cell survival and potentially block apoptosis, thereby facilitating drug resistance [17]. Additionally, stromal cells can modulate tumor cell signaling, survival, proliferation, and drug sensitivity based on the specific microenvironmental context, either conferring resistance or sensitization to various therapeutic agents [18].

Moreover, the interactions between tumor cells and the stromal microenvironment can activate pathways that lead to metabolic adaptations, such as changes in energy metabolism and redox balance, which further support drug resistance [19]. The TME can also induce a protective niche that not only shelters cancer cells from therapeutic agents but also enhances their ability to withstand treatment-induced stresses [20].

Recent studies have highlighted the complexity of these interactions, noting that the tumor microenvironment can also be reshaped in response to therapy, which may exacerbate drug resistance. For instance, during radiotherapy, stromal cells can undergo remodeling that supports tumor progression and therapy resistance [21]. This dynamic response underscores the necessity of understanding the intricate signaling networks within the TME to develop effective therapeutic strategies.

In conclusion, the tumor microenvironment, particularly through the roles of stromal cells, significantly contributes to cancer drug resistance. Understanding these mechanisms is critical for developing novel therapeutic approaches aimed at overcoming resistance and improving clinical outcomes for cancer patients. Targeting the TME, in conjunction with conventional therapies, presents a promising strategy to enhance treatment efficacy and mitigate resistance [[pmid:25588753],[pmid:30680600],[pmid:26845449]].

4.2 Hypoxia and Nutrient Availability

The tumor microenvironment, particularly hypoxia and nutrient availability, plays a crucial role in the development of drug resistance in cancer therapy. Hypoxia, a condition characterized by reduced oxygen availability, is a common feature of solid tumors due to rapid tumor growth and insufficient blood supply. This microenvironmental condition significantly influences the behavior of cancer cells and their response to therapeutic interventions.

Hypoxia induces various adaptive mechanisms in tumor cells, which are critical for their survival and proliferation under stress conditions. One of the primary responses to hypoxia is the activation of hypoxia-inducible factors (HIFs), particularly HIF-1α. HIF-1α regulates the expression of numerous genes involved in angiogenesis, metabolism, and cell survival. For instance, it promotes the production of angiogenic factors that enhance blood vessel formation, allowing tumors to better cope with low oxygen levels [22].

Moreover, hypoxic conditions lead to metabolic reprogramming of cancer cells. Under low oxygen availability, cancer cells often shift their metabolism towards glycolysis, a process that does not require oxygen, to generate energy. This shift allows them to survive in nutrient-poor environments but can also lead to the accumulation of metabolic byproducts, such as lactate, which can further inhibit the effectiveness of therapies [23].

The presence of hypoxia is associated with increased resistance to chemotherapy and radiotherapy. Several mechanisms contribute to this resistance. Firstly, hypoxia can impair the delivery and efficacy of chemotherapeutic agents, as many of these drugs require oxygen to exert their cytotoxic effects. Additionally, hypoxia promotes cellular adaptations that enhance survival, such as the upregulation of ATP-binding cassette (ABC) transporters, which can actively pump out chemotherapeutic drugs from cancer cells [24].

Furthermore, the hypoxic tumor microenvironment can lead to alterations in the immune landscape of the tumor. It is known to promote immune evasion by inducing the polarization of macrophages towards a protumorigenic M2 phenotype and facilitating T cell exhaustion. This immune suppression further complicates the effectiveness of immunotherapies, which rely on an active immune response to eliminate cancer cells [25].

In addition to hypoxia, nutrient availability also significantly impacts drug resistance. Tumor cells often experience nutrient deprivation due to the chaotic and insufficient blood supply. In response, they may utilize alternative metabolic pathways to sustain their growth. For example, under glucose-deficient conditions, hypoxic cancer cells can adapt by utilizing other substrates, such as glutamine and fatty acids, which can support their energy needs and contribute to tumor progression [26].

Overall, the interplay between hypoxia and nutrient availability in the tumor microenvironment creates a challenging landscape for cancer treatment. The adaptations that cancer cells undergo in response to these conditions not only promote their survival but also enhance their resistance to conventional therapies. Understanding these mechanisms is crucial for developing more effective treatment strategies that can overcome the challenges posed by the tumor microenvironment. Targeting the pathways associated with hypoxia and nutrient metabolism may offer promising avenues for improving therapeutic outcomes in cancer patients [27][28].

5 Cancer Stem Cells and Resistance

5.1 Characteristics of Cancer Stem Cells

Cancer drug resistance is a multifaceted challenge that significantly hampers the effectiveness of cancer therapies. One of the critical contributors to this resistance is the presence of cancer stem cells (CSCs), which possess unique characteristics that enable them to survive and thrive even in the presence of therapeutic agents.

CSCs are defined by their ability to self-renew and differentiate into various cell types within a tumor. They are often morphologically and phenotypically distinct from the bulk of tumor cells, exhibiting features that contribute to their resilience against conventional therapies. Notably, CSCs are implicated in the mechanisms of drug resistance due to several inherent properties:

  1. Self-Renewal and Differentiation: CSCs can regenerate the tumor by self-renewing and differentiating into heterogeneous cancer cell populations. This ability allows them to sustain tumor growth even after the bulk of the tumor has been targeted by chemotherapy, leading to cancer relapse (Rezayatmand et al. 2022) [29].

  2. Signaling Pathway Reprogramming: CSCs have been shown to reprogram their signaling mechanisms to promote stemness, which in turn enhances their drug resistance. The activation of specific signaling pathways associated with stemness can lead to an altered response to therapeutic interventions, making it imperative to understand these pathways for the development of effective treatments (Khan et al. 2020) [30].

  3. Epithelial-to-Mesenchymal Transition (EMT): CSCs often undergo EMT, a process that endows them with increased migratory and invasive capabilities, as well as enhanced resistance to apoptosis. This transition is associated with the expression of various drug efflux pumps and detoxification genes, further contributing to their survival in the presence of chemotherapy (Adhikari et al. 2022) [31].

  4. Enhanced DNA Repair Mechanisms: CSCs exhibit a greater capacity for DNA repair compared to non-stem cancer cells. This enhanced repair capability allows them to withstand DNA-damaging agents commonly used in cancer therapies, such as chemotherapy and radiation (Roszkowska 2024) [7].

  5. Microenvironmental Influence: The tumor microenvironment plays a crucial role in promoting CSC survival and drug resistance. Interactions with surrounding stromal cells and the extracellular matrix can provide protective signals that enhance the resistance of CSCs to therapeutic agents (Dalton 2003) [32].

  6. Multidrug Resistance (MDR) Mechanisms: CSCs frequently express high levels of ATP-binding cassette (ABC) transporters, which function as efflux pumps that actively remove therapeutic agents from the cells, thereby reducing drug accumulation and effectiveness (Tu et al. 2022) [33].

In summary, the characteristics of cancer stem cells—such as their ability to self-renew, reprogram signaling pathways, undergo EMT, enhance DNA repair, interact with the tumor microenvironment, and express drug efflux pumps—collectively contribute to the mechanisms of drug resistance in cancer. Understanding these features is critical for developing novel therapeutic strategies aimed at targeting CSCs to improve treatment outcomes and combat drug resistance effectively.

5.2 Mechanisms of Resistance in Cancer Stem Cells

Cancer drug resistance remains a significant challenge in oncology, particularly due to the unique characteristics of cancer stem cells (CSCs). These cells, often identified as a small subset within tumors, exhibit stem-like properties such as self-renewal and differentiation into heterogeneous cancer cell populations. Their resilience to conventional therapies is attributed to several intricate mechanisms.

One of the primary mechanisms of resistance in CSCs involves the expression of specific markers and signaling pathways that facilitate their survival and proliferation. For instance, CSCs often undergo epithelial-to-mesenchymal transition (EMT), which enhances their migratory and invasive capabilities while simultaneously contributing to drug resistance. EMT is characterized by the downregulation of E-cadherin and the upregulation of transcription factors like Snail and Twist, which are known to promote a more aggressive tumor phenotype [34].

Additionally, CSCs possess robust drug efflux mechanisms, primarily mediated by ATP-binding cassette (ABC) transporters. These transporters actively pump out therapeutic agents, reducing drug accumulation within the cells and thereby diminishing the efficacy of treatment [31]. The quiescent nature of many CSCs further complicates therapy, as these cells can evade the cytotoxic effects of drugs that target rapidly dividing cells [29].

The tumor microenvironment also plays a critical role in CSC drug resistance. Interactions with stromal cells and extracellular matrix components can activate signaling pathways that promote cell survival and drug tolerance. For example, cell adhesion-mediated drug resistance (CAM-DR) is a phenomenon where CSCs adhere to the extracellular matrix, activating survival pathways that protect them from apoptosis induced by chemotherapy [17].

Moreover, CSCs are capable of undergoing metabolic reprogramming, allowing them to adapt to various stressors, including chemotherapeutic agents. This metabolic flexibility can enable CSCs to survive in hostile environments, further contributing to their drug-resistant phenotype [35].

The epigenetic landscape of CSCs also significantly influences their resistance mechanisms. Epigenetic modifications can alter gene expression patterns associated with drug metabolism, apoptosis, and DNA repair, thus enhancing the survival of CSCs under therapeutic pressure [31].

Recent studies have highlighted the importance of the DNA damage response (DDR) in CSCs. An enhanced DDR allows these cells to repair drug-induced DNA damage more effectively than their differentiated counterparts, thus contributing to their resistance to chemotherapeutic agents [36].

In summary, the mechanisms of drug resistance in cancer stem cells are multifaceted, involving a combination of cellular, microenvironmental, and epigenetic factors. Understanding these mechanisms is crucial for developing targeted therapies aimed at eradicating CSCs and overcoming resistance in cancer treatment. Addressing the unique features of CSCs may lead to more effective therapeutic strategies and improved patient outcomes [29][31][37].

6 Clinical Implications and Strategies to Overcome Resistance

6.1 Predictive Biomarkers

Cancer drug resistance is a multifaceted phenomenon that poses significant challenges in oncology, affecting treatment efficacy and patient outcomes. The mechanisms underlying this resistance can be broadly categorized into genetic, epigenetic, and microenvironmental factors, each contributing to the reduced responsiveness of cancer cells to therapeutic agents.

Genetic mechanisms of resistance often involve somatic mutations that alter drug targets or affect pathways critical for drug action. For instance, studies have shown that up to 63% of somatic mutations can be heterogeneous within individual tumors, which contributes to the development of resistance (Roszkowska 2024) [7]. Additionally, mutations can lead to changes in drug metabolism, surface drug receptors, and the development of alternative pathways that allow cancer cells to bypass the effects of targeted therapies (Gottesman et al. 2016) [6].

Epigenetic alterations also play a crucial role in cancer drug resistance. These modifications can influence gene expression without changing the underlying DNA sequence, thus enabling tumor cells to adapt to therapeutic pressures. For example, the expression of certain microRNAs, such as miR-34, has been linked to drug resistance mechanisms, where low levels of miR-34 correlate with poor responses to chemotherapy (Naghizadeh et al. 2020) [38].

The tumor microenvironment (TME) significantly impacts drug resistance as well. Components of the TME, including cancer-associated fibroblasts and immune cell populations, can create a supportive niche that enhances tumor survival and promotes resistance (Lei et al. 2023) [3]. Tumor plasticity, which refers to the ability of tumor cells to adapt their phenotype in response to environmental changes, further complicates the resistance landscape (Roszkowska 2024) [7].

To address these challenges, various strategies have been proposed to enhance the effectiveness of cancer treatments and mitigate resistance. One approach involves the identification of predictive biomarkers that can help stratify patients based on their likelihood of responding to specific therapies. This is crucial for personalizing treatment regimens and avoiding ineffective therapies that could harm patients (Tan et al. 2010) [39].

Combination therapies that target multiple signaling pathways simultaneously are another promising strategy. By attacking the tumor from various angles, these approaches can potentially overcome the compensatory mechanisms that cancer cells employ to survive treatment (Vijayakumar et al. 2024) [1]. Moreover, advancements in drug design and delivery systems, including the use of nanoparticles, are being explored to improve the targeting and efficacy of anticancer agents (Lei et al. 2023) [3].

The development of novel therapeutic agents that specifically target the identified resistance mechanisms is also underway. For instance, inhibitors that target specific mutations in DNA repair pathways or signaling cascades have shown promise in preclinical and clinical studies (Sarmento-Ribeiro et al. 2019) [40].

In conclusion, understanding the complex mechanisms of cancer drug resistance is essential for developing effective treatment strategies. By focusing on predictive biomarkers and innovative combination therapies, clinicians can enhance treatment efficacy and improve outcomes for patients facing resistant cancers. The ongoing research in this area continues to hold the potential for significant advancements in personalized cancer therapy.

6.2 Combination Therapies

Cancer drug resistance is a complex phenomenon that significantly hampers the efficacy of cancer therapies, leading to treatment failure and poor patient outcomes. The mechanisms underlying drug resistance are multifaceted and can be broadly categorized into genetic, epigenetic, and microenvironmental factors.

Genetic alterations play a crucial role in drug resistance. These can include mutations in drug targets, amplification of genes associated with drug metabolism, and changes in the expression of transport proteins that affect drug uptake and efflux. For instance, alterations in drug transport mechanisms can lead to reduced intracellular drug accumulation or increased efflux, thus diminishing the therapeutic efficacy of the drugs administered (Zahreddine and Borden, 2013) [41]. Furthermore, tumor heterogeneity can contribute to resistance, where subpopulations of cancer cells may inherently possess or acquire resistance features under selective pressure from treatment (Koirala and DiPaola, 2024) [42].

Epigenetic modifications also contribute to drug resistance by altering gene expression without changing the underlying DNA sequence. These modifications can lead to impaired apoptosis and the activation of survival pathways, allowing cancer cells to withstand therapeutic interventions (Chatterjee and Bivona, 2019) [43]. The tumor microenvironment (TME) plays a significant role in modulating resistance mechanisms, as factors within the TME can influence drug sensitivity and the overall response to treatment (Vijayakumar et al., 2024) [1].

To address the challenge of drug resistance, various strategies have been proposed. One of the most promising approaches is the use of combination therapies. By employing multiple agents with different mechanisms of action, it is possible to target various pathways involved in resistance, thereby enhancing treatment efficacy and preventing the emergence of resistant clones. Combination therapies can block multiple escape pathways essential for tumor survival, which is particularly important in the context of multidrug resistance (Yardley, 2013) [44].

The rational design of combination therapies requires a deep understanding of the molecular mechanisms associated with therapy resistance. This includes identifying biomarkers that can predict drug response and resistance, which can guide the selection of appropriate combinations (Jin et al., 2023) [45]. Additionally, innovative approaches such as targeted protein degradation, immunotherapy combinations, and novel drug delivery systems are being explored to improve treatment outcomes and overcome resistance (Wang et al., 2023) [46].

In conclusion, the mechanisms of cancer drug resistance are diverse and involve a combination of genetic, epigenetic, and microenvironmental factors. Addressing these challenges through combination therapies and a thorough understanding of the underlying resistance mechanisms holds promise for improving cancer treatment outcomes and reducing the incidence of treatment failure.

7 Conclusion

The investigation into cancer drug resistance has revealed a complex interplay of genetic, epigenetic, and microenvironmental factors that contribute to treatment failure. Key findings indicate that genetic mutations in drug targets, activation of alternative signaling pathways, and the role of cancer stem cells significantly enhance the resilience of tumors against therapeutic agents. Additionally, the tumor microenvironment, particularly through stromal interactions and conditions like hypoxia, plays a crucial role in modulating drug responses. Future research directions should focus on the identification of predictive biomarkers, the development of combination therapies targeting multiple resistance mechanisms, and the exploration of innovative therapeutic strategies that can overcome the challenges posed by drug resistance. By addressing these multifaceted aspects, we can improve therapeutic efficacy and patient outcomes in cancer treatment.

References

  • [1] Sudikshaa Vijayakumar;Raveena Dhakshanamoorthy;Akshaya Baskaran;B Sabari Krishnan;Ravi Maddaly. Drug resistance in human cancers - Mechanisms and implications.. Life sciences(IF=5.1). 2024. PMID:39004273. DOI: 10.1016/j.lfs.2024.122907.
  • [2] Tanweer Haider;Vikas Pandey;Nagma Banjare;Prem N Gupta;Vandana Soni. Drug resistance in cancer: mechanisms and tackling strategies.. Pharmacological reports : PR(IF=3.8). 2020. PMID:32700248. DOI: 10.1007/s43440-020-00138-7.
  • [3] Zi-Ning Lei;Qin Tian;Qiu-Xu Teng;John N D Wurpel;Leli Zeng;Yihang Pan;Zhe-Sheng Chen. Understanding and targeting resistance mechanisms in cancer.. MedComm(IF=10.7). 2023. PMID:37229486. DOI: 10.1002/mco2.265.
  • [4] Yun Lei;Shuang Cai;Jia-Kui Zhang;Si-Qi Ding;Zhan-He Zhang;Chun-Dong Zhang;Dong-Qiu Dai;Yong-Shuang Li. The role and mechanism of fatty acid oxidation in cancer drug resistance.. Cell death discovery(IF=7.0). 2025. PMID:40514365. DOI: 10.1038/s41420-025-02554-1.
  • [5] Behzad Mansoori;Ali Mohammadi;Sadaf Davudian;Solmaz Shirjang;Behzad Baradaran. The Different Mechanisms of Cancer Drug Resistance: A Brief Review.. Advanced pharmaceutical bulletin(IF=4.1). 2017. PMID:29071215. DOI: 10.15171/apb.2017.041.
  • [6] Michael M Gottesman;Orit Lavi;Matthew D Hall;Jean-Pierre Gillet. Toward a Better Understanding of the Complexity of Cancer Drug Resistance.. Annual review of pharmacology and toxicology(IF=13.1). 2016. PMID:26514196. DOI: 10.1146/annurev-pharmtox-010715-103111.
  • [7] Malgorzata Roszkowska. Multilevel Mechanisms of Cancer Drug Resistance.. International journal of molecular sciences(IF=4.9). 2024. PMID:39596466. DOI: 10.3390/ijms252212402.
  • [8] Roel H Wilting;Jan-Hermen Dannenberg. Epigenetic mechanisms in tumorigenesis, tumor cell heterogeneity and drug resistance.. Drug resistance updates : reviews and commentaries in antimicrobial and anticancer chemotherapy(IF=21.7). 2012. PMID:22356866. DOI: 10.1016/j.drup.2012.01.008.
  • [9] Nauf Bou Antoun;Athina-Myrto Chioni. Dysregulated Signalling Pathways Driving Anticancer Drug Resistance.. International journal of molecular sciences(IF=4.9). 2023. PMID:37569598. DOI: 10.3390/ijms241512222.
  • [10] Andreas Wicki;Mario Mandalà;Daniela Massi;Daniela Taverna;Huifang Tang;Brian A Hemmings;Gongda Xue. Acquired Resistance to Clinical Cancer Therapy: A Twist in Physiological Signaling.. Physiological reviews(IF=28.7). 2016. PMID:27142452. DOI: 10.1152/physrev.00024.2015.
  • [11] Mariangela Russo. Genetic and non-genetic drug resistance: Darwin or Lamarck?. Molecular oncology(IF=4.5). 2024. PMID:38308461. DOI: 10.1002/1878-0261.13601.
  • [12] Su Jin Choi;Seok Won Jung;Sora Huh;Yoon-Seok Chung;Hyosun Cho;Hyojeung Kang. Alteration of DNA Methylation in Gastric Cancer with Chemotherapy.. Journal of microbiology and biotechnology(IF=3.1). 2017. PMID:28621113. DOI: 10.4014/jmb.1704.04035.
  • [13] Suryendu Saha;Samikshya Mahapatra;Sinjan Khanra;Barnalee Mishra;Biswajit Swain;Diksha Malhotra;Swarnali Saha;Venketesh K Panda;Kavita Kumari;Sarmistha Jena;Sandeep Thakur;Pawan K Singh;Gopal C Kundu. Decoding breast cancer treatment resistance through genetic, epigenetic, and immune-regulatory mechanisms: from molecular insights to translational perspectives.. Cancer drug resistance (Alhambra, Calif.)(IF=5.2). 2025. PMID:40843360. DOI: 10.20517/cdr.2025.69.
  • [14] Lasse Sommer Kristensen;Helene Myrtue Nielsen;Lise Lotte Hansen. Epigenetics and cancer treatment.. European journal of pharmacology(IF=4.7). 2009. PMID:19836388. DOI: 10.1016/j.ejphar.2009.10.011.
  • [15] Nan Wang;Ting Ma;Bin Yu. Targeting epigenetic regulators to overcome drug resistance in cancers.. Signal transduction and targeted therapy(IF=52.7). 2023. PMID:36797239. DOI: 10.1038/s41392-023-01341-7.
  • [16] Hana Q Sadida;Alanoud Abdulla;Sara Al Marzooqi;Sheema Hashem;Muzafar A Macha;Ammira S Al-Shabeeb Akil;Ajaz A Bhat. Epigenetic modifications: Key players in cancer heterogeneity and drug resistance.. Translational oncology(IF=4.1). 2024. PMID:37931371. DOI: 10.1016/j.tranon.2023.101821.
  • [17] William S. Dalton. The tumor microenvironment as a determinant of drug response and resistance.. Drug resistance updates : reviews and commentaries in antimicrobial and anticancer chemotherapy(IF=21.7). 1999. PMID:11504502. DOI: 10.1054/drup.1999.0097.
  • [18] Douglas W McMillin;Joseph M Negri;Constantine S Mitsiades. The role of tumour-stromal interactions in modifying drug response: challenges and opportunities.. Nature reviews. Drug discovery(IF=101.8). 2013. PMID:23449307. DOI: 10.1038/nrd3870.
  • [19] Wan Zhang;Peng Huang. Cancer-stromal interactions: role in cell survival, metabolism and drug sensitivity.. Cancer biology & therapy(IF=4.6). 2011. PMID:21191189. DOI: 10.4161/cbt.11.2.14623.
  • [20] Edna Cukierman;Daniel E Bassi. The mesenchymal tumor microenvironment: a drug-resistant niche.. Cell adhesion & migration(IF=3.5). 2012. PMID:22568991. DOI: 10.4161/cam.20210.
  • [21] Tracy J Berg;Alexander Pietras. Radiotherapy-induced remodeling of the tumor microenvironment by stromal cells.. Seminars in cancer biology(IF=15.7). 2022. PMID:35143991. DOI: 10.1016/j.semcancer.2022.02.011.
  • [22] Jae-Young Kim;Joo-Yong Lee. Targeting Tumor Adaption to Chronic Hypoxia: Implications for Drug Resistance, and How It Can Be Overcome.. International journal of molecular sciences(IF=4.9). 2017. PMID:28841148. DOI: 10.3390/ijms18091854.
  • [23] Wen Cheng;Xian Xiao;Yang Liao;Qingqing Cao;Chaoran Wang;Xiaojiang Li;Yingjie Jia. Conducive target range of breast cancer: Hypoxic tumor microenvironment.. Frontiers in oncology(IF=3.3). 2022. PMID:36226050. DOI: 10.3389/fonc.2022.978276.
  • [24] Jean-Philippe Cosse;Carine Michiels. Tumour hypoxia affects the responsiveness of cancer cells to chemotherapy and promotes cancer progression.. Anti-cancer agents in medicinal chemistry(IF=3.0). 2008. PMID:18855580. DOI: 10.2174/187152008785914798.
  • [25] Xiaoliang Cheng;Peixing Wang;Hongqiang Lyu;Yonghyun Lee;Juyoung Yoon;Haiyan Dong. Targeting the hypoxia signaling pathway with nanomedicine to reverse immunotherapy resistance.. Cancer drug resistance (Alhambra, Calif.)(IF=5.2). 2025. PMID:41019982. DOI: 10.20517/cdr.2025.132.
  • [26] Hamid A Bakshi;Michella Mkhael;Hakkim L Faruck;Asad Ullah Khan;Alaa A A Aljabali;Vijay Mishra;Mohamed El-Tanani;Nitin B Charbe;Murtaza M Tambuwala. Cellular signaling in the hypoxic cancer microenvironment: Implications for drug resistance and therapeutic targeting.. Cellular signalling(IF=3.7). 2024. PMID:37805102. DOI: 10.1016/j.cellsig.2023.110911.
  • [27] Zhou Chen;Fangfang Han;Yan Du;Huaqing Shi;Wence Zhou. Hypoxic microenvironment in cancer: molecular mechanisms and therapeutic interventions.. Signal transduction and targeted therapy(IF=52.7). 2023. PMID:36797231. DOI: 10.1038/s41392-023-01332-8.
  • [28] Raefa Abou Khouzam;Bassam Janji;Jerome Thiery;Rania Faouzi Zaarour;Ali N Chamseddine;Hemma Mayr;Pierre Savagner;Claudine Kieda;Sophie Gad;Stéphanie Buart;Jean-Marie Lehn;Perparim Limani;Salem Chouaib. Hypoxia as a potential inducer of immune tolerance, tumor plasticity and a driver of tumor mutational burden: Impact on cancer immunotherapy.. Seminars in cancer biology(IF=15.7). 2023. PMID:38029865. DOI: 10.1016/j.semcancer.2023.11.008.
  • [29] Hamed Rezayatmand;Mahboobeh Razmkhah;Iman Razeghian-Jahromi. Drug resistance in cancer therapy: the Pandora's Box of cancer stem cells.. Stem cell research & therapy(IF=7.3). 2022. PMID:35505363. DOI: 10.1186/s13287-022-02856-6.
  • [30] Abdul Q Khan;Khalid Rashid;Abdulhadi A AlAmodi;Syed Shadab Raza;Shahab Uddin. Recent developments in unraveling signaling mechanisms underlying drug resistance due to cancer stem-like cells.. Current opinion in pharmacology(IF=4.2). 2020. PMID:33166909. DOI: 10.1016/j.coph.2020.09.012.
  • [31] Swagata Adhikari;Apoorva Bhattacharya;Santanu Adhikary;Vipin Singh;Shrikanth S Gadad;Siddhartha Roy;Chandrima Das. The paradigm of drug resistance in cancer: an epigenetic perspective.. Bioscience reports(IF=4.7). 2022. PMID:35438143. DOI: 10.1042/BSR20211812.
  • [32] William S Dalton. The tumor microenvironment: focus on myeloma.. Cancer treatment reviews(IF=10.5). 2003. PMID:12738239. DOI: 10.1016/s0305-7372(03)00077-x.
  • [33] Shi-Ming Tu;Charles C Guo;Diana S-L Chow;Niki M Zacharias. Stem Cell Theory of Cancer: Implications for Drug Resistance and Chemosensitivity in Cancer Care.. Cancers(IF=4.4). 2022. PMID:35326699. DOI: 10.3390/cancers14061548.
  • [34] Paraskevi Mallini;Thomas Lennard;John Kirby;Annette Meeson. Epithelial-to-mesenchymal transition: what is the impact on breast cancer stem cells and drug resistance.. Cancer treatment reviews(IF=10.5). 2014. PMID:24090504. DOI: .
  • [35] Camille Leonce;Pierre Saintigny;Sandra Ortiz-Cuaran. Cell-Intrinsic Mechanisms of Drug Tolerance to Systemic Therapies in Cancer.. Molecular cancer research : MCR(IF=4.7). 2022. PMID:34389691. DOI: 10.1158/1541-7786.MCR-21-0038.
  • [36] Etna Abad;Dmitry Graifer;Alex Lyakhovich. DNA damage response and resistance of cancer stem cells.. Cancer letters(IF=10.1). 2020. PMID:31968219. DOI: 10.1016/j.canlet.2020.01.008.
  • [37] Huldani Huldani;Saade Abdalkareem Jasim;Klunko Nataliya Sergeenva;Dmitry Olegovich Bokov;Walid Kamal Abdelbasset;Rustam Turakulov;Moaed E Al-Gazally;Behnam Ahmadzadeh;Zanko Hassan Jawhar;Homayoon Siahmansouri. Mechanisms of cancer stem cells drug resistance and the pivotal role of HMGA2.. Pathology, research and practice(IF=3.2). 2022. PMID:35468338. DOI: 10.1016/j.prp.2022.153906.
  • [38] Sanaz Naghizadeh;Ali Mohammadi;Pascal H G Duijf;Behzad Baradaran;Elham Safarzadeh;William Chi-Shing Cho;Behzad Mansoori. The role of miR-34 in cancer drug resistance.. Journal of cellular physiology(IF=4.0). 2020. PMID:32064620. DOI: 10.1002/jcp.29640.
  • [39] Daniel S-W Tan;Marco Gerlinger;Bin-Tean Teh;Charles Swanton. Anti-cancer drug resistance: understanding the mechanisms through the use of integrative genomics and functional RNA interference.. European journal of cancer (Oxford, England : 1990)(IF=7.1). 2010. PMID:20413300. DOI: 10.1016/j.ejca.2010.03.019.
  • [40] Ana Bela Sarmento-Ribeiro;Andreas Scorilas;Ana Cristina Gonçalves;Thomas Efferth;Ioannis P Trougakos. The emergence of drug resistance to targeted cancer therapies: Clinical evidence.. Drug resistance updates : reviews and commentaries in antimicrobial and anticancer chemotherapy(IF=21.7). 2019. PMID:31733611. DOI: 10.1016/j.drup.2019.100646.
  • [41] Hiba Zahreddine;Katherine L B Borden. Mechanisms and insights into drug resistance in cancer.. Frontiers in pharmacology(IF=4.8). 2013. PMID:23504227. DOI: 10.3389/fphar.2013.00028.
  • [42] Mahesh Koirala;Mario DiPaola. Overcoming Cancer Resistance: Strategies and Modalities for Effective Treatment.. Biomedicines(IF=3.9). 2024. PMID:39200265. DOI: 10.3390/biomedicines12081801.
  • [43] Nilanjana Chatterjee;Trever G Bivona. Polytherapy and Targeted Cancer Drug Resistance.. Trends in cancer(IF=17.5). 2019. PMID:30898264. DOI: 10.1016/j.trecan.2019.02.003.
  • [44] Denise A Yardley. Drug resistance and the role of combination chemotherapy in improving patient outcomes.. International journal of breast cancer(IF=3.0). 2013. PMID:23864953. DOI: 10.1155/2013/137414.
  • [45] Haojie Jin;Liqin Wang;René Bernards. Rational combinations of targeted cancer therapies: background, advances and challenges.. Nature reviews. Drug discovery(IF=101.8). 2023. PMID:36509911. DOI: 10.1038/s41573-022-00615-z.
  • [46] Zi'an Wang;Yueqin Wang;Zeyun Li;Wenhua Xue;Shousen Hu;Xiangzhen Kong. Lipid metabolism as a target for cancer drug resistance: progress and prospects.. Frontiers in pharmacology(IF=4.8). 2023. PMID:37841917. DOI: 10.3389/fphar.2023.1274335.

MaltSci Intelligent Research Services

Search for more papers on MaltSci.com

cancer drug resistance mechanisms · genetic mutations · epigenetic modifications · tumor microenvironment · cancer stem cells


© 2025 MaltSci