Preprint
Review

This version is not peer-reviewed.

Molecular Mechanisms of Targeted Therapy Resistance in Genitourinary Tumors: A Path to New Therapeutic Horizons

Submitted:

10 June 2025

Posted:

12 June 2025

You are already at the latest version

Abstract
Targeted therapies have transformed the treatment of genitourinary (GU) malignancies, particularly renal cell carcinoma (RCC), offering significant clinical benefits. However, therapy resistance, driven by intrinsic tumor characteristics and acquired mechanisms, frequently limits their effectiveness. This review comprehensively examines targeted therapies in GU tumors, focusing on their clinical impact and the molecular basis of resistance. While RCC has shown substantial improvements in overall survival, other GU tumors like prostate and bladder cancer have experienced more modest gains due to molecular heterogeneity and complex resistance pathways. These resistance mechanisms are diverse, including genomic alterations, epigenetic modifications, tumor microenvironment influences (immunosuppressive cells, angiogenesis), activation of alternative signaling pathways (PI3K/AKT, MAPK), and metabolic reprogramming. Intratumoral and metastatic site heterogeneity further contribute to resistant subclones. Current strategies to combat resistance involve developing next-generation agents, combination therapies (immunotherapy with TKIs), and personalized medicine guided by genomic profiling and biomarkers like PD-L1, TMB, and ctDNA. Liquid biopsies are increasingly used for real-time monitoring of resistance. Translational research focuses on innovative clinical trial designs (adaptive, basket, umbrella) and real-world evidence. Multidisciplinary collaborations, addressing undertreated populations, and navigating regulatory and cost challenges are crucial. Future directions include breakthroughs in NGS and single-cell analysis, big data and machine learning for predictive modeling, organoids and PDXs for preclinical studies, and novel TME-directed therapies. Ultimately, a more profound understanding of resistance mechanisms and innovative approaches will lead to more effective, personalized, and durable treatments for GU tumors, redefining clinical paradigms and improving patient outcomes.
Keywords: 
;  ;  ;  ;  ;  ;  

Introduction

Genitourinary (GU) malignancies represent a substantial global health challenge, with prostate, bladder, and kidney cancers accounting for the highest incidence and mortality rates within this category [1,2,3,4]. Prostate cancer, for instance, is the second most frequently diagnosed cancer in men worldwide, with over 1.4 million new cases in 2020 and significant mortality, particularly in populations with limited access to early detection [1,2]. Bladder cancer, ranking as the second most common GU malignancy, is responsible for nearly 500,000 new cases annually, whereas kidney cancer (primarily renal cell carcinoma) contributes roughly 400,000 new diagnoses per year, both placing substantial burdens on healthcare systems [3,4]. Disparities in healthcare infrastructure, alongside behavioral and environmental risk factors such as smoking and obesity, exacerbate these statistics, reflecting observed geographical variations in GU cancer outcomes [1,5,6].
Against this epidemiological backdrop, the treatment landscape for GU tumors has undergone a paradigm shift with the rise of targeted therapies. These modalities include agents directed at specific molecular pathways—most notably immune checkpoint inhibitors (ICIs) and targeted small-molecule inhibitors—that have demonstrated durable remission and improved survival in select patient subgroups [7,8]. Immune-based therapies, such as those targeting PD-1 and PD-L1, have significantly impacted the management of advanced renal cell carcinoma and urothelial carcinoma, often being used in combination with anti-angiogenic agents [8,9]. Meanwhile, the introduction of next-generation androgen receptor (AR) inhibitors (e.g., enzalutamide, abiraterone) has redefined the standard of care in metastatic castration-resistant prostate cancer by extending survival and delaying disease progression [9]. In bladder cancer with identifiable fibroblast growth factor receptor (FGFR) alterations, oral FGFR inhibitors such as erdafitinib have offered new avenues for personalized therapy [10,11].
A key force driving these therapeutic successes is the growing knowledge of GU tumor biology. Molecular studies reveal that genetic abnormalities, dysregulated signaling pathways, and complex interactions in the tumor microenvironment collectively shape tumor progression [12,13]. For example, the frequent presence of AR gene mutations in prostate cancer underscores the value of hormonal pathway inhibition, while alterations in FGFR3 in bladder tumors validate the use of FGFR-targeting agents [14,15]. Epigenetic changes—such as DNA methylation, histone modifications, and regulatory noncoding RNAs—further compound the heterogeneity of GU cancers, influencing their susceptibility to various treatments [16,17]. Consequently, research initiatives increasingly focus on biomarker discovery to elucidate which molecular perturbations drive resistance to specific targeted agents [7].
In the past five years, these molecular insights have fueled personalized medicine approaches that tailor therapies based on individual tumor profiles [18]. Advanced genomic tests, circulating tumor DNA (ctDNA) assays, and dedicated molecular tumor boards (MTBs) help guide the selection of targeted treatments for GU malignancies [19]. This patient-centered framework has enhanced response rates and allowed clinicians to adjust therapies in real time as new resistance mechanisms emerge [14,17]. Additionally, immune checkpoint blockade, guided by PD-L1 expression and other biomarkers, underscores the potential of combining molecular profiling with immunotherapeutic strategies for disease subtypes once considered refractory [7].
Despite these encouraging developments, major challenges persist. Tumor heterogeneity remains a formidable barrier, as varying clonal populations and complex microenvironments within the same tumor can foster both intrinsic and acquired resistance [20,21]. In many instances, robust predictive biomarkers are lacking; without validated markers, it is difficult to discern which patients will derive substantial benefit from targeted agents [22]. Further complicating the landscape are adverse effects related to these treatments, including immune-related toxicities that necessitate therapy adjustments and may limit long-term adherence [23]. Access disparities, driven by socioeconomic variables and healthcare infrastructure, also restrict the real-world impact of these innovations, particularly in low- and middle-income regions [24,25]. These limitations highlight the importance of ongoing research to improve biomarker-based interventions, optimize combination regimens, and locate new anticancer targets to overcome resistance.
The aim of this review is to synthesize recent insights into the molecular mechanisms underlying targeted therapy resistance in GU tumors and to highlight emerging strategies designed to overcome these challenges. By integrating key findings from clinical trials, meta-analyses, and novel translational research, we will examine the biological drivers of resistance, identify promising therapeutic approaches (including next-generation targeted agents and combination regimens), and discuss the evolving role of precision medicine. Ultimately, this review seeks to provide a structured framework for clinicians, researchers, and healthcare policymakers to better understand resistance mechanisms, leverage current targeted therapies more effectively. It also aims to guide future investigative directions toward more personalized and durable treatment strategies in GU oncology.

Overview of Targeted Therapies in Gu Tumors

Genitourinary cancers, notably prostate, bladder, and kidney malignancies, are driven by complex webs of signaling cascades and oncogenic processes that promote tumor growth, survival, and spread [26,27,28]. Research characterizing these pathways has shed light on key molecular targets, and this evolving knowledge underpins the development of therapeutic strategies that aim to inhibit crucial elements of tumor cell biology. A key route commonly dysregulated in GU malignancies is the PI3K/AKT/mTOR pathway [26], which, when activated—often due to inactivating mutations in PTEN—drives uncontrolled cell proliferation. In prostate cancer, this dysregulation contributes to disease progression and therapy resistance [28,29]. Clear cell renal cell carcinoma (ccRCC) tends to display similar upregulation of this pathway through loss of the VHL gene, with stabilized hypoxia-inducible factors (HIFs) stimulating mTOR to support angiogenesis [26]. Drugs such as everolimus, which block the mTOR component, have consequently been introduced, particularly in advanced disease settings [28].
In prostate tumors, Wnt/β-catenin signaling also plays a significant role, promoting enhanced cancer stemness and inducing epithelial-mesenchymal transition [30,31]. This process can escalate when androgen signaling is suppressed [32], indicating extensive crosstalk between Wnt activity and the androgen receptor (AR) pathway and underscoring the complexity of the regulatory network in prostate cancer [33]. Notch signaling, in turn, has been tied to the regulation of cancer stem cells that influence tumor recurrence in prostate cancer [34], and Notch inhibition has shown the potential to enhance the effects of chemotherapy such as docetaxel [35].
The TGF-β/Smad signaling cascade, present in both prostate and bladder tumors, features a dual nature by exerting tumor-suppressive effects in early disease but fostering epithelial-mesenchymal transition and metastatic capacity in later stages [36,37]. Beyond these signaling pathways, oncogenic drivers such as EGFR, VEGF/VEGFR, PD-1/PD-L1, and FGFR have received considerable attention in bladder and kidney cancers [38,39,40]. EGFR overexpression correlates with high-grade and advanced bladder tumors [41], whereas VEGF production driven by VHL dysfunction is integral to angiogenesis in ccRCC [40]. Clinical trials validating VEGF and EGFR inhibition illustrate how therapeutics can effectively disrupt these pathways to impede tumor growth [41]. Similarly, PD-1/PD-L1 blockade, shown to be effective in advanced bladder cancer and RCC, underlines the importance of immunoregulatory pathways, while FGFR inhibition addresses FGFR3 alterations common in certain bladder tumor subsets [39].
By mapping these intersecting molecular routes, researchers have moved beyond traditional hormone or chemotherapy toward targeted treatments that more precisely inhibit the main biological drivers of GU cancers. Although these targeted agents demonstrate measurable efficacy, many tumors activate multiple or overlapping pathways that contribute to resistance, prompting ongoing investigations into combination regimens that integrate two or more therapeutic approaches, including immunotherapy [7,14]. The continual refinement of biomarker identification and genomic profiling of GU cancers also improves patient selection, ensuring that those with specific pathway dysregulations or relevant molecular signatures receive the most appropriate targeted therapies. The emerging consensus is that an increasingly nuanced view of GU tumors—and the interactions of their central signaling routes—will guide next-generation trials and clinical practice, ultimately advancing patient outcomes.
The introduction of targeted therapies has significantly altered the management of GU malignancies, yet their clinical impact and associated limitations vary considerably across different tumor types [42,43]. Renal cell carcinoma (RCC) stands out as the GU tumor type that has demonstrated the most significant improvement in overall survival (OS) with targeted agents, largely due to its well-defined molecular pathogenesis involving the VHL gene and angiogenic pathways [44,45,46]. Tyrosine kinase inhibitors (TKIs) and mTOR inhibitors targeting these pathways have translated into marked OS gains, particularly with antiangiogenic agents like sunitinib and pazopanib [44,45,46]. This success is partly attributed to the identification of robust biomarkers in RCC, such as those related to the VHL pathway, which facilitate patient selection [44,47]. Furthermore, the integration of immunotherapy with antiangiogenic agents in RCC has yielded even greater survival benefits, leveraging both direct molecular inhibition and immune modulation [48,49].
In contrast, prostate and bladder cancers have generally lagged behind RCC in terms of OS improvement with targeted therapies [42,43,50]. While agents targeting androgen receptor (AR) signaling in prostate cancer and various pathways in bladder cancer have shown activity, the OS gains have been more modest, often limited to progression-free survival (PFS) endpoints in clinical trials [43,50,51]. This disparity is linked to the greater molecular complexity and heterogeneity of prostate and bladder tumors, which present a broader array of genetic alterations and adaptive resistance mechanisms compared to the more predictable angiogenic dependence of RCC [21,42,50]. Testicular cancer, while a GU malignancy, has historically been highly responsive to conventional chemotherapy, with targeted therapies playing a less significant role in further OS improvement [45,47].
Comparing outcomes from randomized controlled trials (RCTs) and real-world data (RWD) in GU oncology reveals important nuances [52,53]. RCTs, with their stringent selection criteria and controlled environments, frequently demonstrate robust efficacy and safety profiles in relatively homogeneous populations [54]. However, RWD, derived from routine clinical practice, captures a broader spectrum of patients, including those with comorbidities, advanced age, or complex disease presentations typically excluded from trials [53]. Consequently, RWD analyses in mRCC, for example, have sometimes reported more tempered OS outcomes compared to RCTs, reflecting factors such as treatment adherence, comorbidities, and variations in management [54]. While RCTs provide high internal validity, RWD offers essential external validity, highlighting how targeted therapies perform in diverse real-life settings and informing clinical guidelines to better reflect everyday practice [52,53]. Advanced statistical methods are increasingly used to bridge the gap, but differences in data quality and study design persist [52].
Despite the clinical benefits, targeted therapies in GU tumors are associated with a spectrum of adverse events (AEs) and toxicities that can lead to treatment discontinuation or dose reduction [55,56,57,58,59]. Frequently reported AEs include hematologic toxicities (anemia, thrombocytopenia, leukopenia) with VEGFR inhibitors like pazopanib, gastrointestinal disturbances (nausea, diarrhea), and fatigue, which is a common nonhematologic toxicity across many targeted agents [57]. Cutaneous toxicities, ranging from rash to severe reactions like Stevens–Johnson syndrome, are notable with antibody-drug conjugates (ADCs) such as enfortumab vedotin, particularly in combination with immune checkpoint inhibitors [56]. Immune checkpoint inhibitors themselves are associated with immune-related adverse events (irAEs) affecting various organs, including colitis, hepatitis, endocrinopathies, and renal toxicities like acute tubulointerstitial nephritis [57,58]. FGFR inhibitors can cause hyperphosphatemia and stomatitis [59].
Factors predicting treatment discontinuation or dose reduction due to toxicity are multifaceted. Patient-specific characteristics such as advanced age, higher baseline comorbidity burden, and poor performance status are consistently identified predictors [60,61]. Pharmacokinetic variability, prior treatment history, baseline laboratory abnormalities, and concurrent medications also influence tolerance [60,62]. Early onset of severe AEs, high-grade hematologic or cardiovascular toxicities, and inadequate supportive care infrastructure are further predictors [60,61,63]. Patient-reported outcomes and psychological factors like anxiety can also influence decisions to modify or discontinue therapy [64]. Multidisciplinary collaboration and institutional experience in toxicity management can mitigate these risks [63].
Despite the identification of promising predictive factors for targeted therapy benefit, large-scale meta-analyses have not yet conclusively identified specific patient subgroups most likely to benefit across all GU tumors [65,66,67,68,69]. While early tumor shrinkage or certain molecular markers show promise in individual studies or smaller cohorts, heterogeneity in study designs, patient populations, and biomarker assays limits the ability to draw definitive conclusions from pooled analyses. The complexity and molecular diversity of GU tumors, along with the evolving landscape of targeted agents, contribute to this challenge.
Primary reasons for therapy failure or limited duration of response in targeted treatments for GU tumors include intrinsic tumor heterogeneity, acquired resistance mechanisms (such as de novo mutations or epigenetic changes), and the influence of the tumor microenvironment [70,71,72]. Other factors include the induction of EMT, the presence of cancer stem cells, metabolic reprogramming, alterations in drug metabolism and pharmacokinetics, immune evasion, and the activation of alternative signaling pathways [72,73,74,75,76]. Inefficient drug penetration into the tumor and dynamic interactions between tumor and stromal cells also contribute to resistance [70].
Pharmacokinetic (PK) and pharmacodynamic (PD) factors significantly impact targeted therapy efficacy across different GU cancer subtypes [77]. PK variability, influenced by age, organ function, and genetics, affects drug exposure at the tumor site [78,79]. PD factors, such as the degree of target pathway inhibition, are measured through biomarkers and reflect the drug's effect on tumor biology [77]. The interplay of PK and PD, including drug absorption, distribution, metabolism, and excretion, shapes the therapeutic window and influences efficacy and toxicity [77,78]. PK/PD modeling is particularly important for complex agents like ADCs [80].
While complete cures remain rare, long-term follow-up studies have demonstrated durable responses with targeted agents in subsets of patients with advanced GU cancers, particularly with immune checkpoint inhibitors [7,81,82,83,84]. These durable responses, sometimes lasting for years, have translated into significant improvements in OS and quality of life, fundamentally altering the disease trajectory for some patients [7,81,83]. Combination regimens have also shown promise in producing durable responses in rare GU tumor subtypes [85].
The high costs of targeted therapies and variable insurance coverage significantly affect clinical decision-making in GU tumor treatment [64,86,87]. Financial toxicity can lead to delayed treatment initiation, dose reductions, or selection of less expensive, potentially less effective, alternatives [86,88]. Insurance policies and reimbursement frameworks dictate access, with disparities observed across regions and socioeconomic groups [64,86]. Cost-effectiveness evaluations, genomic testing costs, and administrative hurdles like prior authorizations further influence decisions [87,89]. Multidisciplinary tumor boards and financial navigators are increasingly involved to address these challenges [87,90].
Combination regimens with targeted therapies have demonstrated the best outcomes in advanced or metastatic GU cancers, particularly in mRCC and urothelial carcinoma [91,92]. Dual checkpoint inhibition (nivolumab plus ipilimumab) has shown superior OS and response rates compared to monotherapy in intermediate- and poor-risk mRCC [91]. Combinations of VEGF inhibitors with immune checkpoint inhibitors (e.g., axitinib plus pembrolizumab) also improve outcomes by modulating the tumor microenvironment [93]. In urothelial carcinoma, combinations of checkpoint inhibitors with other targeted agents or chemotherapy are being explored to overcome resistance [94]. Rational design, patient selection based on biomarkers, and careful toxicity management are crucial for optimizing these combinations [91,92,93,94].

Definitions and Classifications of Therapy Resistance

Therapy resistance in GU tumors presents a significant challenge in achieving durable clinical responses to targeted agents. This resistance can be broadly categorized into two fundamental types: intrinsic and acquired, each driven by distinct molecular mechanisms and influencing therapeutic outcomes differently. Intrinsic resistance refers to the inherent lack of response to a therapy from the outset, often linked to pre-existing molecular characteristics of the tumor [11,95]. Conversely, acquired resistance develops over time in tumors that initially responded to treatment but subsequently evolve mechanisms to evade its effects [96,97].
The molecular distinctions between intrinsic and acquired resistance are rooted in their underlying genomic and epigenetic alterations. Intrinsic resistance can arise from inherent genomic features such as specific mutations or genomic instability present before therapy initiation [98,99]. For example, mutations in the FGFR3-TACC3 axis in bladder cancer have been associated with primary resistance to FGFR-targeted therapies [98]. Similarly, loss-of-function mutations in JAK1/2 can confer intrinsic resistance to PD-1 blockade by disrupting antigen presentation [100,101]. Epigenetic modifications, including DNA methylation and histone alterations, also contribute to intrinsic resistance by silencing genes involved in drug response or activating survival pathways [102,103]. In contrast, acquired resistance often involves the selection and expansion of subclones with new genomic alterations or adaptive epigenetic changes that emerge under therapeutic pressure [104,105]. These changes can lead to the activation of alternative signaling pathways or the upregulation of immunosuppressive features in the tumor microenvironment (TME) [106,107].
Alternative signaling pathways and compensatory mechanisms are commonly activated in GU tumors to bypass targeted therapy, contributing significantly to both intrinsic and acquired resistance. The PI3K/AKT pathway is a frequent bypass route, activated when upstream targets like FGFR or AR are inhibited, promoting cell survival and proliferation [108,109,110]. The RAS/RAF/MEK/ERK pathway also serves as a compensatory mechanism; for instance, ERBB2/3 upregulation can bypass FGFR inhibition in bladder cancer [111]. Tumors can also engage alternative receptor tyrosine kinases (RTKs) or activate feedback loops to sustain growth signaling despite targeted blockade [112,113,114,115]. The TME further facilitates these bypass mechanisms through paracrine signaling, such as the transfer of resistance signals via extracellular vesicles [116].
The patterns and mechanisms of resistance differ among prostate, bladder, and kidney cancers, reflecting their unique molecular landscapes. Prostate cancer resistance often involves modifications in AR signaling, including mutations and amplifications, alongside epigenetic dysregulation like hypermethylation of tumor suppressor genes [117,118]. Bladder cancer resistance is influenced by genomic instability, mutations in oncogenes like FGFR3 and TP53, and immune evasion mechanisms mediated by PD-L1 expression and the TME [7,119,120,121]. Kidney cancer resistance, particularly in ccRCC, is strongly linked to VHL loss and subsequent dysregulation of hypoxia-mediated pathways, promoting angiogenesis and metabolic changes that confer resistance to VEGF-targeted therapies [119].
Intratumoral and metastatic site heterogeneity significantly contributes to targeted therapy resistance in GU tumors. Intratumoral heterogeneity involves diverse clonal populations with varying genetic alterations and biomarker expression levels within a single tumor [122,123,124]. Metastatic sites can exhibit distinct molecular characteristics and microenvironments compared to the primary tumor, influencing drug response and resistance patterns [125,126,127,128]. This spatial and temporal heterogeneity allows resistant subclones to survive and proliferate under selective pressure [129].
Clonal evolution under targeted therapy pressure leads to the emergence of resistant subclones. Various models describe this process, where pre-existing resistant clones expand or new resistance-conferring mutations arise and are selected for, eventually dominating the tumor population [130,131,132]. Subclonal alterations in driver genes and the dynamic interplay with the TME influence this evolutionary trajectory [133,134,135].
The tumor microenvironment plays a crucial role in mediating resistance and shaping clonal dynamics in GU tumors. Immunosuppressive cells like TAMs, the physical properties of the ECM, and factors like hypoxia, create a protective niche for tumor cells and promote resistance [136,137,138,139]. The TME also exerts selective pressures, favoring the survival and proliferation of resistant clones and influencing the tumor's overall clonal composition [131,140].
Validated and emerging biomarkers (tissue or liquid) are crucial for predicting intrinsic resistance or the likelihood of developing acquired resistance. Established biomarkers include PD-L1 expression and TMB for predicting immunotherapy response, although their predictive value for targeted agents varies [141,142,143]. FGFR alterations are validated predictors for FGFR inhibitor response in urothelial carcinoma [144]. Emerging liquid biopsy biomarkers like ctDNA and CTCs offer non-invasive monitoring of resistance mechanisms and clonal evolution [145,146,147]. Other emerging biomarkers include TIL characteristics, MSI status, and microRNA signatures [148,149].
Liquid biopsies, specifically ctDNA and CTCs, are increasingly used to monitor the emergence of acquired resistance and clonal evolution in real-time. Analyzing ctDNA allows for the detection of resistance-conferring mutations as they arise under therapeutic pressure [150,151]. CTC analysis provides insights into the phenotypic and genetic evolution of tumor cells circulating in the bloodstream [147,152]. Serial liquid biopsies enable dynamic tracking of the tumor's molecular landscape, helping to identify shifts in clonal composition and predict treatment failure before radiographic progression [153,154,155].
Cross-resistance, where resistance to one agent confers resistance to others, is documented through preclinical models and clinical observations [156,157,158]. This phenomenon is often mediated by shared resistance mechanisms, such as the activation of common bypass pathways or the influence of the TME [159,160]. Novel strategies to overcome established resistance mechanisms include targeting the TME (e.g., inhibiting FAP or reprogramming TAMs), enhancing immunotherapy (e.g., dendritic cell activation), inhibiting alternative oncogenic pathways (e.g., targeting HGF/c-MET), and utilizing innovative drug delivery systems like bioresponsive hydrogels [161,162,163,164,165,166,167,168].

Molecular Mechanisms of Resistance

Resistance to targeted therapies in GU tumors is a complex phenomenon driven by a multitude of molecular mechanisms, encompassing genomic alterations, epigenetic modifications, interactions within the TME, activation of alternative signaling pathways, and metabolic adaptations. Understanding these mechanisms is crucial for developing strategies to overcome resistance and improve patient outcomes.
Genomic alterations represent a fundamental driver of targeted therapy resistance in GU tumors. These include frequent mutations, copy number variations (CNVs), and gene fusions [169,170,171]. Alterations in the androgen receptor (AR) and PI3K pathway components are particularly common in prostate cancer, contributing significantly to resistance [169]. Resistance can emerge through mechanisms such as receptor amplification or aberrant activation of downstream signaling pathways driven by mutations [169,172]. Copy number alterations, including gains in loci like MYC, PIK3CA, and CCNE1 have been associated with increased aggressiveness and resistance in various cancers, including GU tumors [173,174]. Furthermore, gene fusions can render targeted therapies ineffective by altering downstream effects or eliminating targetable proteins [171,172].
Alterations in tumor suppressor genes, such as PTEN and TP53, also play a critical role in resistance. Loss of function in TP53 leads to impaired cell cycle control and apoptosis, promoting uncontrolled proliferation and therapeutic escape [175,176,177]. Inactivation of PTEN, a negative regulator of the PI3K/AKT pathway, results in constitutive AKT activation, enhancing cell survival and resistance to therapies targeting this pathway [176,178]. The emergence of AR splice variants, particularly AR-V7, is a significant mechanism of resistance to AR-targeted therapies in prostate cancer, driving androgen-independent growth and bypassing conventional hormonal treatments [179,180]. These variants lack the ligand-binding domain, allowing them to remain active despite low androgen levels, and their presence is associated with poor prognosis and diminished response to agents like enzalutamide and abiraterone [179,181].
Epigenetic modifications, including DNA methylation, histone alterations, and noncoding RNAs, are crucial drivers of resistance phenotypes. Aberrant DNA methylation, particularly hypermethylation of tumor suppressor gene promoters, can silence gene expression and contribute to resistance [182,183]. Histone modifications, such as acetylation and methylation, alter chromatin structure and gene accessibility, influencing the expression of genes involved in drug response [184,185]. Histone deacetylase (HDAC) inhibitors are being explored to reverse these changes and restore sensitivity [186,187]. Noncoding RNAs, including microRNAs and long noncoding RNAs, regulate resistance-related pathways by modulating gene expression, affecting drug metabolism, apoptosis, and proliferation [188,189,190]. Chromatin remodeling and accessibility, influenced by these epigenetic mechanisms, mediate gene expression changes that enable cancer cells to adapt and resist therapy [191,192].
The tumor microenvironment (TME) significantly contributes to therapy resistance and shapes clonal dynamics. Cellular components like tumor-associated macrophages (TAMs) and regulatory T cells (Tregs) create an immunosuppressive environment that hinders antitumor immunity and promotes resistance, including to immunotherapy [136,193,194]. Cancer-associated fibroblasts (CAFs) secrete growth factors and cytokines, remodel the extracellular matrix, and can induce drug efflux pumps, creating a protective niche for tumor cells and enhancing resistance [195,196,197]. TME factors such as cytokines (e.g., IL-6, TGF-β), angiogenesis, and hypoxia promote resistance and metabolic adaptations [194,198,199,200]. Hypoxia induces metabolic reprogramming, shifting cells towards glycolysis (Warburg effect) and activating survival pathways like those driven by HIF-1α, enabling survival under therapeutic stress [201,202,203].
Activation of alternative signaling pathways and compensatory feedback loops is a common mechanism for bypassing targeted therapy. The PI3K/AKT/mTOR pathway is frequently activated upon inhibition of upstream targets, promoting survival and proliferation [204,205,206]. The MAPK pathway (RAS/RAF/MEK/ERK) also serves as an alternative route, with feedback activation allowing tumor cells to escape targeted inhibition [26,207,208]. Crosstalk between key signaling pathways, such as AR signaling and the PI3K/AKT/mTOR or MAPK pathways, facilitates resistance in GU tumors, particularly prostate cancer [26,209,210,211,212]. The interaction between HIF-1α and AR signaling also contributes to resistance under hypoxic conditions [212,213].
Drug efflux pumps, primarily ABC transporters, and broader metabolic reprogramming significantly reduce drug efficacy. Overexpression of ABC transporters like P-glycoprotein actively extrudes drugs from cells, contributing to multidrug resistance [214,215]. Metabolic adaptations, including enhanced glycolysis, altered glutamine and lipid metabolism, and activation of the pentose phosphate pathway, provide alternative energy sources and support cell survival under therapeutic pressure [216,217,218,219,220,221]. The interplay between efflux pumps and metabolic shifts creates a complex resistance landscape [222,223].
Genomic alterations and the TME can differ significantly between primary and metastatic sites, influencing resistance patterns. Metastatic lesions often acquire distinct mutations and CNVs that promote survival in new microenvironments [224,225]. The TME at metastatic sites can vary in fibroblast composition, immune cell populations (e.g., predominance of M2 macrophages), and metabolic characteristics, creating site-specific resistance profiles [196,226,227,228,229]. These differences contribute to increased drug efflux and altered vulnerability to therapies, complicating treatment strategies for advanced disease [230,231].

Strategies to Overcome Therapy Resistance

Overcoming therapy resistance is a critical objective in the management of advanced GU tumors. As tumors develop complex mechanisms to evade treatment, novel strategies focusing on next-generation targeted agents, rational combination therapies, personalized medicine approaches, and emerging experimental modalities are being actively investigated to restore sensitivity and improve patient outcomes.
Novel classes of targeted agents and next-generation inhibitors are being developed to specifically target resistant GU tumors by exploiting their unique molecular vulnerabilities. Research into bromodomain and extraterminal (BET) inhibitors, particularly for tumors with NUTM1 rearrangements, has shown promise in preclinical models, targeting chromatin interactions crucial in these resistant subtypes [232,233,234]. Next-generation sequencing (NGS) plays a pivotal role by identifying actionable genetic alterations, such as microsatellite instability (MSI) or DNA mismatch repair (dMMR) deficiencies, which predict response to immune checkpoint inhibitors like pembrolizumab [235,236,237]. Emerging RNA-based therapies, including those involving long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), are also being explored as potential biomarkers and therapeutic targets to influence tumor behavior and resistance [238,239]. Additionally, innovative modalities like photothermal and photodynamic therapy (PDT) using nanoparticles are being investigated for their ability to induce localized damage in resistant tumors [240,241].
Combination therapy approaches have shown significant promise in overcoming resistance in advanced GU cancers by targeting multiple pathways simultaneously. Combinations of targeted agents with immunotherapy, such as enfortumab vedotin (an ADC targeting Nectin-4) with pembrolizumab (a PD-1 inhibitor) in metastatic urothelial carcinoma, leverage synergistic effects to counteract immune evasion [7,242]. In castration-resistant prostate cancer, combining androgen receptor inhibitors like enzalutamide with immunotherapeutic approaches has demonstrated efficacy by targeting both androgen signaling and immune responses [243,244,245]. Similarly, combining multi-tyrosine kinase inhibitors like cabozantinib with nivolumab has shown favorable outcomes in advanced renal cell carcinoma [246,247]. Combinations of targeted agents with chemotherapy, such as docetaxel with abiraterone or enzalutamide, are also explored, sometimes employing strategic cycling to exploit treatment-induced vulnerabilities [243,248].
Personalized medicine strategies, including genomic profiling and biomarker identification, are essential for selecting patients for resistance-overcoming therapies. Comprehensive genomic profiling via NGS identifies actionable mutations and alterations, enabling tailored treatment plans based on individual tumor characteristics [249,250,251]. Liquid biopsies, analyzing circulating tumor DNA (ctDNA) and circulating tumor cells (CTCs), provide real-time monitoring of tumor dynamics and emerging resistance mutations, guiding adaptive treatment decisions [18,252,253]. Biomarkers like high tumor mutational burden (TMB) or specific mutations (e.g., TP53, FGFR3) predict response to checkpoint inhibitors and targeted therapies [253]. Patient-derived xenograft (PDX) models also aid in testing therapies against a patient's unique tumor profile [254].
Promising emerging and experimental therapies are being investigated to address resistance mechanisms. Cell therapies, such as CAR T cells targeting PSMA in prostate cancer, aim to overcome resistance associated with AR signaling [14,235]. Gene editing technologies like CRISPR/Cas9 are used to identify and potentially modify genes linked to resistance or target metabolic vulnerabilities in cancer stem cells [255]. Radioligand therapies, such as ^177Lu-PSMA-617, deliver targeted radiation to resistant cells, particularly in castration-resistant prostate cancer [256]. Strategies targeting the TME, including inhibiting STAT3 signaling or modulating hypoxia, aim to reduce tumor resilience and sensitize tumors to existing treatments [257,258,259,260]. Experimental combinations, like PARP inhibitors with immunotherapy or autophagy inhibitors, are also explored [261,262].
Next-generation targeted agents and combination regimens are designed to counteract specific resistance mechanisms, such as bypass pathways or AR splice variants. To address bypass pathways, strategies include dual kinase inhibitors targeting primary drivers and compensatory routes [263,264], multi-kinase inhibitors, and approaches manipulating ROS levels [26,265]. For AR splice variants like AR-V7, next-generation antiandrogens and combinations targeting alternative pathways like the glucocorticoid receptor (GR) are being developed to restore sensitivity [266,267,268]. Combinations targeting AR interactions with co-factors or upstream pathways like FGF and MAPK are also explored [269,270].
Strategies targeting the tumor microenvironment (TME) enhance targeted therapy efficacy and overcome resistance. Targeting tumor-associated fibroblasts (TAFs) can disrupt their pro-tumorigenic and immunosuppressive roles [271,272]. Modulating hypoxia, for example, through improved oxygenation or hypoxia-activated prodrugs, can enhance drug activity [259,273,274]. Immune modulation within the TME, using checkpoint inhibitors or targeting immunosuppressive cells like Tregs and TAMs, promotes antitumor immunity [275,276]. ECM remodeling strategies can improve drug penetration [277,278]. Combination approaches addressing TME factors alongside tumor cells aim to prevent clonal expansion and enhance responses [279,280,281,282,283].
Liquid biopsies and dynamic biomarker monitoring play a crucial role in guiding adaptive treatment strategies to manage evolving resistance. Analyzing ctDNA and CTCs provides real-time insights into tumor evolution and the emergence of resistance mutations [284,285,286,287,288,289,290]. This allows for timely treatment adjustments based on the dynamic molecular landscape [285,286,289,290,291]. Sequential monitoring of biomarkers like ctDNA levels or circulating miRNAs can indicate therapeutic effectiveness or failure and inform adaptive strategies [289,292,293,294].
Epigenetic therapies and other novel modalities are being explored, alone or in combination, to reverse resistance phenotypes. DNA methylation inhibitors (e.g., azacitidine, decitabine) reverse aberrant methylation, reactivating silenced tumor suppressor genes [295,296,297]. HDAC inhibitors alter histone acetylation to promote gene expression [298,299]. Combining epigenetic therapies with immunotherapy can enhance tumor immunogenicity [189,300]. These approaches also target specific resistance mechanisms, such as reactivating silenced tumor suppressor genes or reprogramming cancer stem cells [297,301,302,303].
Key considerations and challenges in designing clinical trials for resistant GU tumors include selecting appropriate, often heterogeneous, patient populations based on biomarkers and prior treatments [304,305,306,307]. Trial design must accommodate adaptive methodologies and carefully evaluate combination therapies, considering sequencing, timing, and potential interactions [308,309,310]. Integrating biomarkers and liquid biopsies for dynamic monitoring requires standardization and validation [311,312]. Understanding TME-mediated resistance and temporal changes in resistance mechanisms is also crucial for trial design [313,314,315,316].
Translational challenges and future directions for integrating these strategies into routine clinical practice involve standardizing liquid biopsy technologies and interpreting complex biomarker changes [145,317,318]. Addressing clinical heterogeneity and navigating regulatory and economic considerations for novel agents and biomarkers are also critical [319]. Education for healthcare providers on interpreting emerging data and managing complex regimens is essential [318,320,321]. Future directions include integrating multi-modal biomarkers, utilizing AI and big data for decision support, and employing innovative trial designs to accelerate clinical translation [145,322,323].

Clinical Implications and Translational Perspectives

Translating the growing understanding of therapy resistance mechanisms in GU tumors into clinical practice is paramount for improving patient outcomes. This process involves integrating the latest research findings into clinical trial designs, leveraging validated and emerging biomarkers for personalized therapy, utilizing real-world evidence, fostering multidisciplinary collaboration, navigating regulatory landscapes, and addressing cost-effectiveness considerations.
Latest research findings on resistance mechanisms are profoundly influencing clinical trial designs for GU tumors. Insights into intrinsic and extrinsic factors contributing to resistance, such as mutations affecting antigen presentation or immunosuppressive cells in the TME, are guiding the development of innovative methodologies [324]. Biomarker-driven approaches, utilizing exploratory biomarkers and non-invasive tools like circulating tumor DNA (ctDNA), are essential for stratifying patients and tailoring therapies based on individual tumor profiles [7,18]. This understanding also promotes the design of rational combination therapies to anticipate and mitigate resistance pathways, as seen in studies exploring combinations based on transcriptomic analyses of resistant pathways like FGFR [325,326]. Innovative trial designs, such as basket trials, are gaining traction to test drugs across multiple tumor types based on specific genetic alterations [327,328].
Validated biomarkers guide personalized therapies in routine clinical practice for GU oncology patients. Tumor mutational burden (TMB) and microsatellite instability (MSI)/mismatch repair deficiency (dMMR) status are established predictors of response to immune checkpoint inhibitors (ICIs) in GU cancers [7,141,329,330]. Circulating tumor DNA (ctDNA) is an emerging non-invasive biomarker for monitoring treatment responses, detecting relapses, and identifying actionable mutations that guide targeted therapies [18,331]. While PD-L1 expression remains crucial for patient stratification, its predictive power is enhanced when combined with other biomarkers [141,330]. Gene expression profiles, including lncRNAs and genes involved in DNA repair or EMT, are also being explored as potential biomarkers to inform treatment choices and combination strategies [332,333].
Liquid biopsy technology is being harnessed to guide therapy modifications and monitor resistance in real time. By analyzing ctDNA and circulating tumor cells (CTCs), clinicians can assess treatment efficacy and detect emerging resistance mechanisms, such as acquired mutations, allowing for prompt therapy adjustments [334,335,336,337]. This continuous monitoring provides insights into tumor evolution and guides adaptive treatment strategies based on the dynamic molecular landscape [324,338]. While challenges remain in standardizing methodologies and interpreting results, liquid biopsy facilitates personalized care by providing real-time information on tumor dynamics and responsiveness.
Real-world evidence (RWE) and patient registries inform clinical decision-making about therapy resistance by providing insights beyond controlled clinical trials. RWE captures data from diverse populations in routine practice, revealing how therapies perform across various demographics and treatment contexts [339,340]. Patient registries track long-term outcomes and treatment patterns, highlighting common resistance patterns in broader populations [341]. RWE helps address knowledge gaps in underrepresented subgroups and enhances the external validity of clinical trial findings, supporting evidence-based decision-making [340,342].
Certain GU tumor subpopulations remain undertreated or understudied in clinical trials, including those of advanced age, with significant comorbidities, racial and ethnic minorities, and individuals facing sociodemographic and geographic barriers [1,343,344,345,346]. Adolescents and young adults also face unique challenges and disparities in care [347]. These disparities highlight the need for more inclusive clinical trial designs and tailored approaches to ensure equitable access to advanced therapies and address resistance in these specific populations [307,343].
Multidisciplinary approaches involving pathologists, oncologists, molecular biologists, and bioinformaticians have proven highly effective in tackling resistance. Molecular tumor boards integrate complex genomic data with clinical insights to personalize treatment plans and identify alternative therapies for resistant tumors [348,349]. Pathologists contribute by identifying relevant histopathological features and genetic alterations, while bioinformaticians analyze complex datasets to inform predictive models [349,350]. Collaboration among these experts, along with the use of advanced imaging techniques, facilitates a comprehensive understanding of tumor biology and guides individualized treatment protocols [351,352,353].
Collaborative models exist between academia, industry, and regulatory agencies to accelerate translational research in GU cancers. Cooperative research networks like EORTC facilitate large-scale clinical trials [354]. Innovative frameworks like Pfizer's Centers for Therapeutic Innovation promote hybrid partnerships [355]. Multi-stakeholder platforms like CAREFOR enhance academic clinical trials and bridge communication gaps [356]. Regulatory agencies establish frameworks and guidelines, streamlining approval processes [357]. These collaborations, often integrating RWE, are pivotal in understanding resistance and accelerating novel therapy development [358,359].
Regulatory hurdles can hinder the rapid integration of novel assays or targeted agents into clinical use. Complex regulatory frameworks, differences across regions, and the evolving nature of therapies pose challenges [360,361,362]. Demonstrating value and efficacy, especially for combination therapies, requires robust trial designs [362]. Manufacturing and quality control for novel modalities also present unique hurdles [363]. Regulatory pathways need to be adaptable to accommodate emerging therapies, particularly for rare diseases and precision medicine approaches [364,365].
Cost-effectiveness considerations are crucial when deploying novel targeted or combination therapies. High costs and variable insurance coverage significantly affect clinical decision-making, influencing treatment choice and patient access [366,367,368,369,370]. Cost-effectiveness analyses, considering efficacy, safety, and quality of life, are essential for justifying the financial burden [366,368](Castellano et al., 2022; Benjamin et al., 2024). RWE informs these analyses by providing data on real-world costs and outcomes [369].
Consensus statements and international collaborations push forward new standards in GU tumor care. International consensus recommendations establish standardized outcome measures [371]. Global statistics like GLOBOCAN inform strategies [372]. Organizations like ESMO and ANZUP develop evidence-based guidelines and promote collaborative research [373,374]. These efforts foster uniformity in treatment protocols and accelerate the integration of novel therapies into clinical practice.

Future Directions

The landscape of GU oncology is continuously evolving, driven by a more profound understanding of tumor biology and the mechanisms underlying therapy resistance. Future directions in this field are poised to revolutionize diagnosis, treatment, and patient management through technological advancements, innovative therapeutic strategies, and enhanced collaborative models.
Significant breakthroughs in next-generation sequencing (NGS) and single-cell analysis are on the horizon for mapping resistance phenomena. Single-cell technologies, such as single-cell RNA sequencing (scRNA-seq) and multidimensional mass cytometry, are crucial for uncovering the heterogeneity within tumors and identifying resistant subpopulations that bulk sequencing might miss [375,376,377,378,379,380]. These methods allow for granular analysis of tumor progression, drug responses, and the identification of cellular states contributing to resistance [377,381]. Integrated bioinformatics tools are essential for analyzing the complex data generated, deciphering resistance mechanisms, and identifying potential targets [382,383]. Advancements in targeted NGS and digital microfluidics are also providing insights into resistance in microbial pathogens, highlighting the broader applicability of these technologies [384,385,386]. Emerging single-cell metabolomics will further enhance our understanding of metabolic adaptations in resistant cells, integrating genomic, transcriptomic, and metabolomic data for biomarker discovery [383,387,388,389].
Big data and machine learning (ML) approaches will refine our predictive models for resistance in GU oncology. Leveraging vast datasets from multiomics sources, ML can identify complex patterns and biomarkers predictive of disease progression and treatment responses [390,391]. Integrating circulating tumor DNA (ctDNA) profiling into these models shows promise for predicting responses to immunotherapy and identifying hypermutated environments [392,393]. ML accelerates the identification and validation of novel biomarkers from high-dimensional data [14,394]. Circulating tumor cell (CTC) analysis also enhances predictive models by providing real-time insights into tumor evolution and resistance [395].
Future clinical trial designs, such as adaptive, basket, and umbrella trials, will accelerate novel therapy testing in GU tumors. Adaptive trials offer flexibility to modify protocols based on interim results, accelerating the identification of effective therapies [396,397]. Umbrella trials test multiple therapies targeting different biomarkers within a single disease type, while basket trials assess a single treatment across various tumor types sharing a common molecular feature [327,398,399,400]. Biomarker integration enhances the predictive power of these designs, stratifying patients based on molecular profiles [327,397]. Regulatory bodies are increasingly supportive of these innovative designs to expedite drug development [248,401].
Organoids and patient-derived xenografts (PDXs) hold significant potential for preclinical identification of resistance mechanisms. Patient-derived organoids (PDOs) closely mimic patient tumors, allowing for personalized drug sensitivity testing and elucidation of resistance mechanisms related to specific mutations or adaptations [402,403,404,405]. PDX/organoid platforms capture genomic and phenotypic heterogeneity, facilitating preclinical therapeutic investigations [406]. These models enable the combination of genomic profiling with phenotypic analyses to identify and validate new resistance biomarkers [403,407]. Ultimately, organoids and PDXs support personalized medicine by allowing functional drug testing that replicates patient-specific tumor biology [408].
TME-directed therapies are expected to evolve to encompass more holistic control of immunosuppression and angiogenesis. Targeting hypoxia, for instance, through HIF inhibitors or hypoxia-activated prodrugs, can reduce immunosuppression and enhance therapy efficacy [409,410,411,412]. Modulating tumor-associated macrophages (TAMs) to a pro-inflammatory phenotype is crucial [412,413]. Combination therapies targeting both angiogenic pathways and immune responses, such as VEGF inhibitors with immune checkpoint inhibitors, can normalize tumor vasculature and improve immune cell infiltration [414,415,416,417]. Innovative drug delivery systems using nanoparticles or extracellular vesicles (EVs) can enhance the precision of TME-directed therapies and modulate TME components [418,419,420,421,422,423].
Cross-disciplinary partnerships are needed to push GU tumor research into truly personalized or “precision” territory. Interdisciplinary collaboration among basic scientists, oncologists, and clinical researchers is vital for translating laboratory findings into clinical applications [424,425]. Partnerships with bioinformaticians and data scientists are essential for analyzing large datasets and developing predictive models [426]. Patient-engaged research provides valuable insights into patient needs and preferences [427,428]. Global and public health collaborations enhance the generalizability of findings and address disparities [429,430,431]. Collaboration with regulatory bodies and policymakers streamlines the process of bringing new therapies to market [432]. Translational research consortia facilitate synergistic efforts across institutions [433].
Advanced genomic editing or personalized cell-based therapies face ethical and logistical challenges. Ethical concerns include informed consent, patient autonomy, the potential for eugenics with germline editing, and unintended consequences [434,435,436,437,438,439]. Logistical challenges involve navigating complex regulatory frameworks, ensuring technical standardization and reproducibility, managing long-term monitoring and follow-up, and integrating these innovative therapies into existing healthcare systems [363,364,365,440,441]. Addressing these challenges requires collaboration among stakeholders to ensure responsible application and equitable access.
“Drug holiday” protocols or “dynamic dosing” strategies show potential success in delaying the onset of resistance by reducing selective pressure or adjusting dosages based on tumor response [442,443,444,445]. These approaches align with precision medicine principles, tailoring treatment based on individual patient and tumor characteristics.

Conclusions

The landscape of targeted therapy in GU oncology is characterized by significant advancements and ongoing challenges, particularly concerning the emergence of therapy resistance. A major consensus in the literature is that resistance mechanisms are multifaceted, deeply rooted in the TME, immune evasion strategies, and metabolic reprogramming of cancer cells [7,446,447,448,449,450,451,452,453]. The TME, with its complex interplay of CAFs, TAMs, and other immune cells, actively facilitates resistance by creating an immunosuppressive niche and promoting immune evasion [7,447,448]. Metabolic reprogramming further enables tumor cells to adapt and survive under therapeutic pressure, often impairing immune cell function and contributing to treatment failure [450,452,453]. Additionally, the inherent plasticity of tumor cells, driven by genetic and epigenetic alterations, allows for adaptive resistance mechanisms to emerge, necessitating continuous monitoring and dynamic treatment strategies [454,455].
Several pressing unanswered questions require further investigation to fully validate new therapeutic strategies. A more profound understanding of TME-mediated resistance, particularly the specific interactions between CAFs and TAMs and how they modulate immune responses, is crucial [456]. Reliable biomarkers for predicting patient responses to combination therapies, especially those involving metabolic inhibitors, are needed to guide patient stratification [70,453]. Addressing on-target, off-tumor toxicity of therapies targeting common tumor antigens remains a significant hurdle, requiring the identification of more tumor-specific targets or structural variants [119,457]. The role of epigenetic modulation in enhancing treatment responses, particularly with agents like PARP inhibitors, and the impact of the gut microbiome on therapy efficacy and toxicity also warrant further exploration [458,459,460,461].
Insights into these resistance mechanisms have reshaped the philosophical and clinical approach to treating GU cancers. There is a clear shift from traditional single-agent therapies towards more personalized, combination-based strategies that aim to circumvent the multifaceted nature of resistance [462,463]. Recognizing tumor heterogeneity and the dynamic evolution of resistance has led to the development of adaptive treatment strategies, including the use of liquid biopsies for real-time monitoring of tumor genomics [70,464,465,466,467,468]. Combination therapies targeting multiple pathways simultaneously are increasingly explored to reduce the likelihood of resistance development [469,470,471].
Leading experts offer several recommendations for the future of targeted therapy in GU tumors. Precision medicine, guided by genomic and molecular profiling, is paramount for tailoring individualized treatment plans [14]. Targeting TAMs and other TME components, improving biomarker discovery for patient selection, and considering factors like circadian rhythms in treatment regimens are key areas of focus [141,472,473]. Multi-histology clinical trials and the development of innovative therapeutic agents targeting specific pathways, such as FGFR3, are encouraged [95,474]. Furthermore, strengthening supportive care and fostering interdisciplinary collaborations are vital for advancing GU cancer treatment and addressing complex resistance mechanisms [119].
Finally, healthcare systems must better integrate new evidence on resistance mechanisms into standard treatment guidelines. This involves regularly updating guidelines based on emerging research, implementing multidisciplinary tumor boards, incorporating validated biomarkers into clinical pathways, utilizing clinical decision support tools, encouraging participation in clinical trials focused on resistance, educating and involving patients in shared decision-making, and fostering research collaborations [475,476,477,478,479,480,481,482,483]. Balancing evidence-based decisions with patient-centered values and preferences, including economic considerations and quality of life, is crucial for optimizing care [484,485,486,487,488]. Strategies such as “drug holidays” or “dynamic dosing” are also being explored to delay the onset of resistance by managing selective pressure [489,490,491,492,493,494,495,496,497].
In conclusion, the future of targeted therapy in GU oncology lies in a holistic, personalized, and adaptive approach. By addressing the complexities of resistance through innovative research, collaborative efforts, and patient-centered care, the field aims to significantly improve outcomes for patients with genitourinary malignancies.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer J Clin. 2021, 71, 209–49. [Google Scholar] [CrossRef] [PubMed]
  2. Rawla, P. Epidemiology of Prostate Cancer. World J Oncol. 2019, 10, 63–89. [Google Scholar] [CrossRef] [PubMed]
  3. Ridyard, D.G.; Buller, D.M.; Ristau, B.T. The Current State of Adjuvant Therapy Following Surgery for High-risk Renal Cell Carcinoma. Eur Urol Focus. 2019, 5, 935–8. [Google Scholar] [CrossRef] [PubMed]
  4. Xiao, L.; Xu, C.; Lin, P.; Mu, L.; Yang, X. Novel Dihydroartemisinin Derivative Mito-DHA5 Induces Apoptosis Associated with Mitochondrial Pathway in Bladder Cancer Cells. 2021. [Google Scholar]
  5. Ferlay, J.; Colombet, M.; Soerjomataram, I.; Parkin, D.M.; Piñeros, M.; Znaor, A.; et al. Cancer statistics for the year 2020: An overview. Int J Cancer. 2021, 149, 778–89. [Google Scholar] [CrossRef]
  6. Feng, Z.; Wang, B. Long non-coding RNA HNF1A-AS1 promotes cell viability and migration in human bladder cancer. Oncol Lett. 2018, 15, 4535–40. [Google Scholar] [CrossRef]
  7. Labadie, B.W.; Balar, A.V.; Luke, J.J. Immune Checkpoint Inhibitors for Genitourinary Cancers: Treatment Indications, Investigational Approaches and Biomarkers. Cancers. 2021, 13, 5415. [Google Scholar] [CrossRef]
  8. Schmidt, A.L.; Siefker-Radtke, A.; McConkey, D.; McGregor, B. Renal Cell and Urothelial Carcinoma: Biomarkers for New Treatments. Am Soc Clin Oncol Educ Book. 2020, 40, e197–206. [Google Scholar] [CrossRef]
  9. Maughan, B.L.; Bailey, E.; Gill, D.M.; Agarwal, N. Incidence of Immune-Related Adverse Events with Program Death Receptor-1- and Program Death Receptor-1 Ligand-Directed Therapies in Genitourinary Cancers. Front Oncol. 2017, 7, 56. [Google Scholar] [CrossRef]
  10. Bahleda, R.; Italiano, A.; Hierro, C.; Mita, A.; Cervantes, A.; Chan, N.; et al. Multicenter Phase I Study of Erdafitinib (JNJ-42756493), Oral Pan-Fibroblast Growth Factor Receptor Inhibitor, in Patients with Advanced or Refractory Solid Tumors. Clin Cancer Res. 2019, 25, 4888–97. [Google Scholar] [CrossRef]
  11. Olah, C.; Shmorhun, O.; Klamminger, G.G.; Rawitzer, J.; Sichward, L.; Hadaschik, B.; et al. Immunohistochemistry-based molecular subtypes of urothelial carcinoma derive different survival benefit from platinum chemotherapy. J Pathol: Clin Res. 2025, 11, e70017. [Google Scholar] [CrossRef]
  12. Boguslawska, J.; Kryst, P.; Poletajew, S.; Piekielko-Witkowska, A. TGF-β and microRNA Interplay in Genitourinary Cancers. Cells. 2019, 8, 1619. [Google Scholar] [CrossRef] [PubMed]
  13. Giunchi, F.; Franceschini, T.; Fiorentino, M. A narrative review of individualized treatments of genitourinary tumors: is the future brighter with molecular evaluations? Transl Androl Urol. 10, 1553561–1551561. [CrossRef] [PubMed]
  14. Zarrabi, K.; Paroya, A.; Wu, S. Emerging therapeutic agents for genitourinary cancers. J Hematol Oncol. 2019, 12, 89. [Google Scholar] [CrossRef]
  15. Weyerer, V.; Eckstein, M.; Compérat, E.; Juette, H.; Gaisa, N.T.; Allory, Y.; et al. Pure Large Nested Variant of Urothelial Carcinoma (LNUC) Is the Prototype of an FGFR3 Mutated Aggressive Urothelial Carcinoma with Luminal-Papillary Phenotype. Cancers. 2020, 12, 763. [Google Scholar] [CrossRef]
  16. Ding, L.; Wang, R.; Shen, D.; Cheng, S.; Wang, H.; Lu, Z.; et al. Role of noncoding RNA in drug resistance of prostate cancer. Cell Death Dis. 2021, 12, 590. [Google Scholar] [CrossRef]
  17. Wu S, Mu C, Sun J jia, Hu X rong, Yao Y hong. Role of Exosomal Non-Coding RNA in the Tumour Microenvironment of Genitourinary System Tumours. Technol Cancer Res Treat. 2023, 22, 15330338231198348. [Google Scholar] [CrossRef]
  18. Gerke, M.B.; Jansen, C.S.; Bilen, M.A. Circulating Tumor DNA in Genitourinary Cancers: Detection, Prognostics, and Therapeutic Implications. Cancers. 2024, 16, 2280. [Google Scholar] [CrossRef] [PubMed]
  19. Michaelis, J.; Himmelsbach, R.; Metzger, P.; Lassmann, S.; Börries, M.; Werner, M.; et al. Primary Results of Patients with Genitourinary Malignancies Presented at a Molecular Tumor Board. Urol Int. 2024, 108, 383–91. [Google Scholar] [CrossRef]
  20. Du, X.; Shao, Y.; Gao, H.; Zhang, X.; Zhang, H.; Ban, Y.; et al. CMTR1-ALK: an ALK fusion in a patient with no response to ALK inhibitor crizotinib. Cancer Biol Ther. 2018, 19, 962–6. [Google Scholar] [CrossRef]
  21. Barth, D.A.; Juracek, J.; Slaby, O.; Pichler, M.; Calin, G.A. lncRNA and Mechanisms of Drug Resistance in Cancers of the Genitourinary System. Cancers. 2020, 12, 2148. [Google Scholar] [CrossRef]
  22. Landmesser, M.E.; Raup-Konsavage, W.M.; Lehman, H.L.; Stairs, D.B. Loss of p120ctn causes EGFR-targeted therapy resistance and failure. PLoS ONE. 2020, 15, e0241299. [Google Scholar] [CrossRef] [PubMed]
  23. Sousa, M.; Peate, M.; Lewis, C.; Jarvis, S.; Willis, A.; Hickey, M.; et al. Exploring knowledge, attitudes and experience of genitourinary symptoms in women with early breast cancer on adjuvant endocrine therapy. Eur J Cancer Care. 2018, 27, e12820. [Google Scholar] [CrossRef] [PubMed]
  24. Aziz, Z.; Naseer, H.; Altaf, A. Challenges in Access to New Therapeutic Agents: Marginalized Patients With Cancer in Pakistan and the Need for New Guidelines. JCO Glob Oncol. 2022, 8, e2100132. [Google Scholar] [CrossRef]
  25. Faraj, K.S.; Kaufman, S.R.; Oerline, M.; Dall, C.; Srivastava, A.; Caram, M.E.V.; et al. The 340B Drug Pricing Program and Management of Advanced Prostate Cancer. Cancer Med. 2024, 14, e70552. [Google Scholar] [CrossRef]
  26. Shorning, B.Y.; Dass, M.S.; Smalley, M.J.; Pearson, H.B. The PI3K-AKT-mTOR Pathway and Prostate Cancer: At the Crossroads of AR, MAPK, and WNT Signaling. Int J Mol Sci. 2020, 21, 4507. [Google Scholar] [CrossRef]
  27. GUOL; LIUY; DINGZ; SUNW; YUANM Signal transduction by M3 muscarinic acetylcholine receptor in prostate cancer. Oncol Lett. 2015, 11, 385–92.
  28. Gasmi, A.; Roubaud, G.; Dariane, C.; Barret, E.; Beauval, J.B.; Brureau, L.; et al. Overview of the Development and Use of Akt Inhibitors in Prostate Cancer. J Clin Med. 2021, 11, 160. [Google Scholar] [CrossRef] [PubMed]
  29. Crumbaker, M.; Khoja, L.; Joshua, A. AR Signaling and the PI3K Pathway in Prostate Cancer. Cancers. 2017, 9, 34. [Google Scholar] [CrossRef]
  30. Zhang, X.; Li, H.; Wang, Y.; Zhao, H.; Wang, Z.; Chan, F.L. Nuclear receptor NURR1 functions to promote stemness and epithelial-mesenchymal transition in prostate cancer via its targeting of Wnt/β-catenin signaling pathway. Cell Death Dis. 2024, 15, 234. [Google Scholar] [CrossRef]
  31. Murillo-Garzón, V.; Kypta, R. WNT signalling in prostate cancer. Nat Rev Urol. 2017, 14, 683–96. [Google Scholar] [CrossRef]
  32. Luo, J.; Wang, D.; Wan, X.; Xu, Y.; Lu, Y.; Kong, Z.; et al. Crosstalk Between AR and Wnt Signaling Promotes Castration-Resistant Prostate Cancer Growth. OncoTargets Ther. 2020, 13, 9257–67. [Google Scholar] [CrossRef] [PubMed]
  33. Khurana, N.; Sikka, S.C. Interplay Between SOX9, Wnt/β-Catenin and Androgen Receptor Signaling in Castration-Resistant Prostate Cancer. Int J Mol Sci. 2019, 20, 2066. [Google Scholar] [CrossRef] [PubMed]
  34. Mourkioti, I.; Angelopoulou, A.; Belogiannis, K.; Lagopati, N.; Potamianos, S.; Kyrodimos, E.; et al. Interplay of Developmental Hippo–Notch Signaling Pathways with the DNA Damage Response in Prostate Cancer. Cells. 2022, 11, 2449. [Google Scholar] [CrossRef]
  35. Wang, L.; Zi, H.; Luo, Y.; Liu, T.; Zheng, H.; Xie, C.; et al. Inhibition of Notch pathway enhances the anti-tumor effect of docetaxel in prostate cancer stem-like cells. Stem Cell Res Ther. 2020, 11, 258. [Google Scholar] [CrossRef] [PubMed]
  36. Zhao, B.; Lu, Y.L.; Yang, Y.; Hu, L.B.; Bai, Y.; Li, R.Q.; et al. Overexpression of lncRNA ANRIL promoted the proliferation and migration of prostate cancer cells via regulating let-7a/TGF- β 1/ Smad signaling pathway. Cancer Biomark. 2018, 21, 613–20. [Google Scholar] [CrossRef]
  37. Datta, D.; Aftabuddin Md Gupta, D.K.; Raha, S.; Sen, P. Human Prostate Cancer Hallmarks Map. Sci Rep. 2016, 6, 30691. [Google Scholar] [CrossRef]
  38. Li, W.; Wang, Y.; Tan, S.; Rao, Q.; Zhu, T.; Huang, G.; et al. Overexpression of Epidermal Growth Factor Receptor (EGFR) and HER-2 in Bladder Carcinoma and Its Association with Patients’ Clinical Features. Méd Sci Monit : Int Méd J Exp Clin Res. 2018, 24, 7178–85. [Google Scholar] [CrossRef]
  39. Xie, W.; Chen, F.; Zhang, L.; Lin, B.; Ye, J.; Yu, Z.; et al. Gefitinib effectively treated advanced lung cancer with a rare EGFR L747P mutation in a kidney transplant recipient: the first case report. 2023. [Google Scholar]
  40. Rinaldi, L.; Chiuso, F.; Senatore, E.; Borzacchiello, D.; Lignitto, L.; Iannucci, R.; et al. Downregulation of praja2 restrains endocytosis and boosts tyrosine kinase receptors in kidney cancer. Commun Biol. 2024, 7, 208. [Google Scholar] [CrossRef]
  41. Zubair, T.; Bandyopadhyay, D. Small Molecule EGFR Inhibitors as Anti-Cancer Agents: Discovery, Mechanisms of Action, and Opportunities. Int J Mol Sci. 2023, 24, 2651. [Google Scholar] [CrossRef]
  42. Ghosh, T.M.; Mitra, A.K.; Davis, J.; Cummings, B.; Yates, C.; Arnold, R. Abstract LB-267: Transcriptomic and epigenomic analysis of metastatic castration-resistant prostate cancer and a pan-cancer analysis of its genetic signatures. Cancer Res. 2019;79(13_Supplement):LB-267-LB-267.
  43. Benjamin, D.J.; Rezazadeh, A. Characterization of genitourinary drug approvals by the FDA, 2020-2024. J Clin Oncol. 2025;43(5_suppl):872–872.
  44. Froehner, M.; Hakenberg, O.W.; Wirth, M.P. Molecular Therapy in Urologic Oncology. Urol Int. 2007, 79, 1–7. [Google Scholar] [CrossRef]
  45. Mooney, D.; Paluri, R.; Mehta, A.; Goyal, J.; Sonpavde, G. Update in Systemic Therapy of Urologic Malignancies. Postgrad Med. 2014, 126, 44–54. [Google Scholar] [CrossRef] [PubMed]
  46. Fitzpatrick, J.; Muneer, A.; Rosette Jde la Powles, T. Oxford Textbook of Oncology. 2016;602–27.
  47. Cimadamore, A.; Scarpelli, M.; Santoni, M.; Massari, F.; Tartari, F.; Cerqueti, R.; et al. Genitourinary Tumors: Update on Molecular Biomarkers for Diagnosis, Prognosis and Prediction of Response to Therapy. Curr Drug Metab. 2019, 20, 305–12. [Google Scholar] [CrossRef]
  48. Gandhy, S.U.; Madan, R.A.; Aragon-Ching, J.B. The immunotherapy revolution in genitourinary malignancies. Immunotherapy. 2020, 12, 819–31. [Google Scholar] [CrossRef]
  49. Haidl, F.; Pfister, D.; Heidenreich, A.; Heidegger, I. Antiangiogenic therapies in urogenital malignancies. memo - Mag Eur Méd Oncol. 2017, 10, 202–5. [Google Scholar] [CrossRef] [PubMed]
  50. Sio, T.T.; Ko, J.; Gudena, V.K.; Verma, N.; Chaudhary, U.B. Chemotherapeutic and targeted biological agents for metastatic bladder cancer: A comprehensive review. Int J Urol. 2014, 21, 630–7. [Google Scholar] [CrossRef]
  51. Yim, A.; Alberto, M.; Herold, M.; Woon, D.; Ischia, J.; Bolton, D. “Pass the Genetic Scalpel”: A Comprehensive Review of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) in Urological Cancers. Société Int dUrol J. 2024, 5, 16–30. [Google Scholar] [CrossRef]
  52. Blonde, L.; Khunti, K.; Harris, S.B.; Meizinger, C.; Skolnik, N.S. Interpretation and Impact of Real-World Clinical Data for the Practicing Clinician. Adv Ther. 2018, 35, 1763–74. [Google Scholar] [CrossRef] [PubMed]
  53. Maio, M.D.; Perrone, F.; Conte, P. Real-World Evidence in Oncology: Opportunities and Limitations. Oncol. 2019, 25, e746–52. [Google Scholar] [CrossRef]
  54. Graham, J.; Heng, D.Y. Real-world evidence in metastatic renal cell carcinoma. Tumori J. 2018, 104, 76–82. [Google Scholar] [CrossRef]
  55. Pili, R.; Qin, R.; Flynn, P.J.; Picus, J.; Millward, M.; Ho, W.M.; et al. A Phase II Safety and Efficacy Study of the Vascular Endothelial Growth Factor Receptor Tyrosine Kinase Inhibitor Pazopanib in Patients With Metastatic Urothelial Cancer. Clin Genitourin Cancer. 2013, 11, 477–83. [Google Scholar] [CrossRef]
  56. Müller, B.; Curatolo, R.; Juratli, H.; Husic, A.; Nehring, J.; Potlukova, E.; et al. Severe cutaneous toxicity in a 67-year-old patient with metastatic urothelial carcinoma undergoing therapy with enfortumab vedotin and pembrolizumab. Eur J Case Rep Intern Med. 2024, 11, 005003. [Google Scholar] [CrossRef]
  57. Brahmer, J.R.; Lacchetti, C.; Schneider, B.J.; Atkins, M.B.; Brassil, K.J.; Caterino, J.M.; et al. Management of Immune-Related Adverse Events in Patients Treated With Immune Checkpoint Inhibitor Therapy: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol. 2018, 36, JCO. [Google Scholar] [CrossRef] [PubMed]
  58. Xipell, M.; Victoria, I.; Hoffmann, V.; Villarreal, J.; García-Herrera, A.; Reig, O.; et al. Acute tubulointerstitial nephritis associated with atezolizumab, an anti-programmed death-ligand 1 (pd-l1) antibody therapy. OncoImmunology. 2018, 7, e1445952. [Google Scholar] [CrossRef]
  59. Ertl, I.E.; Shariat, S.F.; Mostafaei, H.; Ilijazi, D.; Loriot, Y. Fibroblast growth factor receptors across urothelial carcinoma landscape. Curr Opin Urol. 2020, 30, 557–65. [Google Scholar] [CrossRef] [PubMed]
  60. Kaymakcalan, M.D.; Xie, W.; Albiges, L.; North, S.A.; Kollmannsberger, C.K.; Smoragiewicz, M.; et al. Risk factors and model for predicting toxicity-related treatment discontinuation in patients with metastatic renal cell carcinoma treated with vascular endothelial growth factor–targeted therapy: Results from the International Metastatic Renal Cell Carcinoma Database Consortium. Cancer. 2015, 122, 411–9. [Google Scholar]
  61. Donskov, F.; Motzer, R.J.; Voog, E.; Hovey, E.; Grüllich, C.; Nott, L.M.; et al. Outcomes based on age in the phase III METEOR trial of cabozantinib versus everolimus in patients with advanced renal cell carcinoma. Eur J Cancer. 2020, 126, 1–10. [Google Scholar] [CrossRef] [PubMed]
  62. Klümpen, H.J.; Samer, C.F.; Mathijssen, R.H.J.; Schellens, J.H.M.; Gurney, H. Moving towards dose individualization of tyrosine kinase inhibitors. Cancer Treat Rev. 2011, 37, 251–60. [Google Scholar] [CrossRef]
  63. Ruiz, J.N.; Belum, V.R.; Creel, P.; Cohn, A.; Ewer, M.; Lacouture, M.E. Current Practices in the Management of Adverse Events Associated With Targeted Therapies for Advanced Renal Cell Carcinoma: A National Survey of Oncologists. Clin Genitourin Cancer. 2014, 12, 341–7. [Google Scholar] [CrossRef]
  64. Yabroff, K.R.; Shi, K.S.; Zhao, J.; Freedman, A.N.; Zheng, Z.; Nogueira, L.; et al. Importance of Patient Health Insurance Coverage and Out-of-Pocket Costs for Genomic Testing in Oncologists’ Treatment Decisions. JCO Oncol Pr. 2024, 20, 429–37. [Google Scholar] [CrossRef]
  65. Krajewski, K.M.; Franchetti, Y.; Nishino, M.; Fay, A.P.; Ramaiya, N.; Abbeele ADVden et, a.l. 10% Tumor Diameter Shrinkage on the First Follow-Up Computed Tomography Predicts Clinical Outcome in Patients With Advanced Renal Cell Carcinoma Treated With Angiogenesis Inhibitors: A Follow-Up Validation Study. Oncol. 2014, 19, 507–14. [Google Scholar] [CrossRef]
  66. Yoshii, Y.; Furukawa, T.; Oyama, N.; Hasegawa, Y.; Kiyono, Y.; Nishii, R.; et al. Fatty Acid Synthase Is a Key Target in Multiple Essential Tumor Functions of Prostate Cancer: Uptake of Radiolabeled Acetate as a Predictor of the Targeted Therapy Outcome. PLoS ONE. 2013, 8, e64570. [Google Scholar] [CrossRef]
  67. Sobol, R.E.; Menander, K.B.; Chada, S.; Wiederhold, D.; Sellman, B.; Talbott, M.; et al. Meta-Analysis of Adenoviral p53 Gene Therapy Clinical Trials in Recurrent Head and Neck Squamous Cell Carcinoma. medRxiv. 2021;2021.01.06.20248743.
  68. Lv, H.; Zhou, Q.H.; Zhong, D.S. A pooled analysis of molecularly targeted agents for treatment of metastatic oesophago-gastric cancer in elderly patients. Arch Méd Sci : AMS. 2020, 16, 253–9. [Google Scholar] [CrossRef] [PubMed]
  69. He, X.; Zhang, Y.; Ma, Y.; Zhou, T.; Zhang, J.; Hong, S.; et al. Optimal tumor shrinkage predicts long-term outcome in advanced nonsmall cell lung cancer (NSCLC) treated with target therapy. Medicine. 2016, 95, e4176. [Google Scholar] [CrossRef] [PubMed]
  70. Kalemoglu, E.; Jani, Y.; Canaslan, K.; Bilen, M.A. The role of immunotherapy in targeting tumor microenvironment in genitourinary cancers. Front Immunol. 2025, 16, 1506278. [Google Scholar] [CrossRef]
  71. Ni, T.; Liu, G.; Huo, T.; Shi, W.; Gu, Q.; Ji, Q. Abstract 6009: Developing drug-induced resistant tumor models for efficacy evaluation of next-generation anticancer therapies. Cancer Res. 2022;82(12_Supplement):6009–6009.
  72. Ramos, P.; Bentires-Alj, M. Mechanism-based cancer therapy: resistance to therapy, therapy for resistance. Oncogene. 2014, 34, 3617–26. [Google Scholar] [CrossRef]
  73. Ni, T.; Zhang, Z.; Tang, X.; Shi, W.; Gu, Q.; Ji, Q. Abstract 2958: Drug induced resistant tumor models enable the development of next-generation anticancer therapeutics. Cancer Res. 2021;81(13_Supplement):2958–2958.
  74. Gumusay, O.; Vitiello, P.P.; Wabl, C.; Corcoran, R.B.; Bardelli, A.; Rugo, H.S. Strategic Combinations to Prevent and Overcome Resistance to Targeted Therapies in Oncology. Am Soc Clin Oncol Educ Book. 2020, 40, e292–308. [Google Scholar] [CrossRef]
  75. Ou, X.; Gao, G.; Habaz, I.A.; Wang, Y. Mechanisms of resistance to tyrosine kinase inhibitor-targeted therapy and overcoming strategies. MedComm. 2024, 5, e694. [Google Scholar] [CrossRef]
  76. Hopper-Borge, E.A.; Nasto, R.E.; Ratushny, V.; Weiner, L.M.; Golemis, E.A.; Astsaturov, I. Mechanisms of tumor resistance to EGFR-targeted therapies. Expert Opin Ther Targets. 2009, 13, 339–62. [Google Scholar] [CrossRef] [PubMed]
  77. Ong, M.; Banerji, U. Oxford Textbook of Oncology. 2016;209–19.
  78. Evans, W.E.; Johnson, J.A. PHARMACOGENOMICS: The Inherited Basis for Interindividual Differences in Drug Response. Annu Rev Genom Hum Genet. 2001, 2, 9–39. [Google Scholar] [CrossRef]
  79. Hurria, A.; Lichtman, S.M. Pharmacokinetics of Chemotherapy in the Older Patient. Cancer Control. 2007, 14, 32–43. [Google Scholar] [CrossRef]
  80. Hedrich, W.D.; Fandy, T.E.; Ashour, H.M.; Wang, H.; Hassan, H.E. Antibody–Drug Conjugates: Pharmacokinetic/Pharmacodynamic Modeling, Preclinical Characterization, Clinical Studies, and Lessons Learned. Clin Pharmacokinet. 2017, 57, 687–703. [Google Scholar] [CrossRef] [PubMed]
  81. Bellmunt, J.; Wit Rde Vaughn, D.J.; Fradet, Y.; Lee, J.L.; Fong, L.; et al. Pembrolizumab as Second-Line Therapy for Advanced Urothelial Carcinoma. N Engl J Med. 2017, 376, 1015–26. [Google Scholar] [CrossRef] [PubMed]
  82. Hansen, A.R.; Massard, C.; Ott, P.A.; Haas, N.B.; Lopez, J.S.; Ejadi, S.; et al. Pembrolizumab for advanced prostate adenocarcinoma: findings of the KEYNOTE-028 study. Ann Oncol. 2018, 29, 1807–13. [Google Scholar] [CrossRef] [PubMed]
  83. Motzer, R.J.; Escudier, B.; McDermott, D.F.; George, S.; Hammers, H.J.; Srinivas, S.; et al. Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. N Engl J Med. 2015, 373, 1803–13. [Google Scholar] [CrossRef]
  84. Massard, C.; Gordon, M.S.; Sharma, S.; Rafii, S.; Wainberg, Z.A.; Luke, J.; et al. Safety and Efficacy of Durvalumab (MEDI4736), an Anti–Programmed Cell Death Ligand-1 Immune Checkpoint Inhibitor, in Patients With Advanced Urothelial Bladder Cancer. J Clin Oncol. 2016, 34, 3119–25. [Google Scholar] [CrossRef]
  85. McGregor, B.A.; Campbell, M.T.; Xie, W.; Farah, S.; Bilen, M.A.; Schmidt, A.L.; et al. Results of a multicenter, phase 2 study of nivolumab and ipilimumab for patients with advanced rare genitourinary malignancies. Cancer. 2020, 127, 840–9. [Google Scholar] [CrossRef]
  86. Shih, Y.C.T.; Xu, Y.; Liu, L.; Smieliauskas, F. Rising Prices of Targeted Oral Anticancer Medications and Associated Financial Burden on Medicare Beneficiaries. J Clin Oncol. 2017, 35, JCO. [Google Scholar] [CrossRef]
  87. Yabroff, K.R.; Zhao, J.; Moor JSde Sineshaw, H.M.; Freedman, A.N.; Zheng, Z.; et al. Factors Associated With Oncologist Discussions of the Costs of Genomic Testing and Related Treatments. JNCI: J Natl Cancer Inst. 2019, 112, 498–506. [Google Scholar] [CrossRef]
  88. Zafar, S.Y.; Peppercorn, J.M.; Schrag, D.; Taylor, D.H.; Goetzinger, A.M.; Zhong, X.; et al. The Financial Toxicity of Cancer Treatment: A Pilot Study Assessing Out-of-Pocket Expenses and the Insured Cancer Patient’s Experience. Oncol. 2013, 18, 381–90. [Google Scholar] [CrossRef]
  89. Gong, J.; Pan, K.; Fakih, M.; Pal, S.; Salgia, R. Value-based genomics. Oncotarget. 2018, 9, 15792–815. [Google Scholar] [CrossRef]
  90. Liu, A.; Vicenzi, P.; Sharma, I.; Orr, K.; Teller, C.; Koentz, M.; et al. Molecular Tumor Boards: The Next Step towards Precision Therapy in Cancer Care. Hematol Rep. 2023, 15, 244–55. [Google Scholar] [CrossRef] [PubMed]
  91. Motzer, R.J.; Tannir, N.M.; McDermott, D.F.; Frontera, O.A.; Melichar, B.; Choueiri, T.K.; et al. Nivolumab plus Ipilimumab versus Sunitinib in Advanced Renal-Cell Carcinoma. N Engl J Med. 2018, 378, 1277–90. [Google Scholar] [CrossRef] [PubMed]
  92. Dudani, S.; Graham, J.; Wells, J.C.; Bakouny, Z.; Pal, S.K.; Dizman, N.; et al. First-line Immuno-Oncology Combination Therapies in Metastatic Renal-cell Carcinoma: Results from the International Metastatic Renal-cell Carcinoma Database Consortium. Eur Urol. 2019, 76, 861–7. [Google Scholar] [CrossRef]
  93. Yi, M.; Zheng, X.; Niu, M.; Zhu, S.; Ge, H.; Wu, K. Combination strategies with PD-1/PD-L1 blockade: current advances and future directions. Mol Cancer. 2022, 21, 28. [Google Scholar] [CrossRef]
  94. Schulz, G.B.; Black, P.C. Combination therapies involving checkpoint-inhibitors for treatment of urothelial carcinoma: a narrative review. Transl Androl Urol. 2021, 0, 0–0. [Google Scholar] [CrossRef] [PubMed]
  95. Kamoun, A.; Reyniès Ade Allory, Y.; Sjödahl, G.; Robertson, A.G.; Seiler, R.; et al. A Consensus Molecular Classification of Muscle-invasive Bladder Cancer. Eur Urol. 2020, 77, 420–33. [Google Scholar] [CrossRef]
  96. Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science. 2018, 359, 1350–5. [Google Scholar] [CrossRef]
  97. Kirtane, K.; Elmariah, H.; Chung, C.H.; Abate-Daga, D. Adoptive cellular therapy in solid tumor malignancies: review of the literature and challenges ahead. J Immunother Cancer. 2021, 9, e002723. [Google Scholar] [CrossRef]
  98. Tong, Z.; Yan, C.; Dong, Y.A.; Yao, M.; Zhang, H.; Liu, L.; et al. Whole-exome sequencing reveals potential mechanisms of drug resistance to FGFR3-TACC3 targeted therapy and subsequent drug selection: towards a personalized medicine. BMC Méd Genom. 2020, 13, 138. [Google Scholar] [CrossRef]
  99. Almassalha, L.M.; Bauer, G.M.; Chandler, J.E.; Gladstein, S.; Szleifer, I.; Roy, H.K.; et al. The Greater Genomic Landscape: The Heterogeneous Evolution of Cancer. Cancer Res. 2016, 76, 5605–9. [Google Scholar] [CrossRef]
  100. Shin, D.S.; Zaretsky, J.M.; Escuin-Ordinas, H.; Garcia-Diaz, A.; Hu-Lieskovan, S.; Kalbasi, A.; et al. Primary Resistance to PD-1 Blockade Mediated by JAK1/2 Mutations. Cancer Discov. 2017, 7, 188–201. [Google Scholar] [CrossRef]
  101. Gurjao, C.; Liu, D.; Hofree, M.; AlDubayan, S.H.; Wakiro, I.; Su, M.J.; et al. Intrinsic Resistance to Immune Checkpoint Blockade in a Mismatch Repair–Deficient Colorectal Cancer. Cancer Immunol Res. 2019, 7, 1230–6. [Google Scholar] [CrossRef] [PubMed]
  102. Tie, Y.; Chen, C.; Yang, Y.; Qian, Z.; Yuan, H.; Wang, H.; et al. Upregulation of let-7f-5p promotes chemotherapeutic resistance in colorectal cancer by directly repressing several pro-apoptotic proteins. Oncol Lett. 2018, 15, 8695–702. [Google Scholar] [CrossRef] [PubMed]
  103. Rieth, J.; Subramanian, S. Mechanisms of Intrinsic Tumor Resistance to Immunotherapy. Int J Mol Sci. 2018, 19, 1340. [Google Scholar] [CrossRef]
  104. Zhang, G.; Frederick, D.T.; Wu, L.; Wei, Z.; Krepler, C.; Srinivasan, S.; et al. Targeting mitochondrial biogenesis to overcome drug resistance to MAPK inhibitors. J Clin Investig. 2016, 126, 1834–56. [Google Scholar] [CrossRef]
  105. França, G.S.; Baron, M.; Pour, M.; King, B.R.; Rao, A.; Misirlioglu, S.; et al. Drug-induced adaptation along a resistance continuum in cancer cells. bioRxiv. 4968. [Google Scholar]
  106. Zhao, X.; Wangmo, D.; Robertson, M.; Subramanian, S. Acquired Resistance to Immune Checkpoint Blockade Therapies. Cancers. 2020, 12, 1161. [Google Scholar] [CrossRef] [PubMed]
  107. Seliger, B.; Massa, C. Modulation of Lymphocyte Functions in the Microenvironment by Tumor Oncogenic Pathways. Front Immunol. 2022, 13, 883639. [Google Scholar] [CrossRef]
  108. Adelaiye-Ogala, R.M.; Gryder, B.; Nguyen, Y.T.M.; Alilin, A.N.; Grayson, A.; Jansson, K.H.; et al. Targeting the PI3K/AKT pathway overcomes enzalutamide resistance by inhibiting induction of the glucocorticoid receptor. bioRxiv. 3803. [Google Scholar]
  109. Datta, J.; Damodaran, S.; Parks, H.; Ocrainiciuc, C.; Miya, J.; Yu, L.; et al. Akt Activation Mediates Acquired Resistance to Fibroblast Growth Factor Receptor Inhibitor BGJ398. Mol Cancer Ther. 2017, 16, 614–24. [Google Scholar] [CrossRef]
  110. Lau, D.K.; Jenkins, L.; Weickhardt, A. Mechanisms of acquired resistance to fibroblast growth factor receptor targeted therapy. Cancer Drug Resist. 2019, 2, 568–79. [Google Scholar] [CrossRef]
  111. Bockorny, B.; Rusan, M.; Chen, W.; Liao, R.G.; Li, Y.; Piccioni, F.; et al. RAS-MAPK reactivation facilitates acquired resistance in FGFR1-amplified lung cancer and underlies a rationale for upfront FGFR-MEK blockade. Mol Cancer Ther. 2018, 17, molcanther. [Google Scholar] [CrossRef]
  112. Ulanet, D.B.; Ludwig, D.L.; Kahn, C.R.; Hanahan, D. Insulin receptor functionally enhances multistage tumor progression and conveys intrinsic resistance to IGF-1R targeted therapy. Proc Natl Acad Sci. 2010, 107, 10791–8. [Google Scholar] [CrossRef] [PubMed]
  113. Buck, E.; Gokhale, P.C.; Koujak, S.; Brown, E.; Eyzaguirre, A.; Tao, N.; et al. Compensatory Insulin Receptor (IR) Activation on Inhibition of Insulin-Like Growth Factor-1 Receptor (IGF-1R): Rationale for Cotargeting IGF-1R and IR in Cancer. Mol Cancer Ther. 2010, 9, 2652–64. [Google Scholar] [CrossRef]
  114. Niederst, M.J.; Engelman, J.A. Bypass Mechanisms of Resistance to Receptor Tyrosine Kinase Inhibition in Lung Cancer. Sci Signal. 2013, 6, re6. [Google Scholar] [CrossRef]
  115. Manstein Vvon Yang, C.M.; Richter, D.; Delis, N.; Vafaizadeh, V.; Groner, B. Resistance of Cancer Cells to Targeted Therapies Through the Activation of Compensating Signaling Loops. Curr Signal Transduct Ther. 2014, 8, 193–202. [Google Scholar] [CrossRef] [PubMed]
  116. Maleki, S.; Jabalee, J.; Garnis, C. The Role of Extracellular Vesicles in Mediating Resistance to Anticancer Therapies. Int J Mol Sci. 2021, 22, 4166. [Google Scholar] [CrossRef]
  117. Izzo, S.; Naponelli, V.; Bettuzzi, S. Flavonoids as Epigenetic Modulators for Prostate Cancer Prevention. Nutrients. 2020, 12, 1010. [Google Scholar] [CrossRef] [PubMed]
  118. Li, L.C.; Carroll, P.R.; Dahiya, R. Epigenetic Changes in Prostate Cancer: Implication for Diagnosis and Treatment. J Natl Cancer Inst. 2005, 97, 103–15. [Google Scholar] [CrossRef]
  119. Alkhouli, M.A.; Bazargan, S.; Pilon-Thomas, S.; Poch, M.; Chahoud, J. Current State of Cell Therapies for Genitourinary Malignancies. Cancer J. 2022, 28, 294–300. [Google Scholar] [CrossRef]
  120. Bernardo, C.; Eriksson, P.; Marzouka, N.; Liedberg, F.; Sjödahl, G.; Höglund, M. Molecular pathology of the luminal class of urothelial tumors. J Pathol. 2019, 249, 308–18. [Google Scholar] [CrossRef]
  121. Arabi, T.Z.; Ashraf, N.; Sabbah, B.N.; Ouban, A. Claudins in genitourinary tract neoplasms: mechanisms, prognosis, and therapeutic prospects. Front Cell Dev Biol. 2023, 11, 1308082. [Google Scholar] [CrossRef]
  122. Zhang, X.; Nguyen, K.D.; Rudnick, P.; Roper, N.; Kawaler, E.; Maity, T.K.; et al. Quantitative mass spectrometry to interrogate proteomic heterogeneity in metastatic lung adenocarcinoma and validate a novel somatic mutation CDK12-G879V. bioRxiv. 8313. [Google Scholar]
  123. Jilaveanu, L.B.; Shuch, B.; Zito, C.R.; Parisi, F.; Barr, M.; Kluger, Y.; et al. PD-L1 Expression in Clear Cell Renal Cell Carcinoma: An Analysis of Nephrectomy and Sites of Metastases. J Cancer. 2014, 5, 166–72. [Google Scholar] [CrossRef] [PubMed]
  124. Saito, T.; Kondo, C.; Shitara, K.; Ito, Y.; Saito, N.; Ikehara, Y.; et al. Comparison of intratumoral heterogeneity of HER2 expression between primary tumor and multiple organ metastases in gastric cancer: Clinicopathological study of three autopsy cases and one resected case. Pathol Int. 2015, 65, 309–17. [Google Scholar] [CrossRef]
  125. Shuch, B.; Falbo, R.; Parisi, F.; Adeniran, A.; Kluger, Y.; Kluger, H.M.; et al. MET Expression in Primary and Metastatic Clear Cell Renal Cell Carcinoma: Implications of Correlative Biomarker Assessment to MET Pathway Inhibitors. BioMed Res Int. 2015, 2015, 192406. [Google Scholar] [CrossRef] [PubMed]
  126. Shi, Y.; Tsang, J.Y.S.; Ni, Y.; Tse, G.M. Intratumoral Heterogeneity in Breast Cancer: A Comparison of Primary and Metastatic Breast Cancers. Oncol. 2017, 22, 487–90. [Google Scholar] [CrossRef] [PubMed]
  127. Grinda, T.; Joyon, N.; Lusque, A.; Lefèvre, S.; Arnould, L.; Penault-Llorca, F.; et al. Phenotypic discordance between primary and metastatic breast cancer in the large-scale real-life multicenter French ESME cohort. npj Breast Cancer. 2021, 7, 41. [Google Scholar] [CrossRef]
  128. Wang, W.; Ye, L.F.; Bao, H.; Hu, M.T.; Han, M.; Tang, H.M.; et al. Heterogeneity and evolution of tumour immune microenvironment in metastatic gastroesophageal adenocarcinoma. Gastric Cancer. 2022, 25, 1017–30. [Google Scholar] [CrossRef]
  129. Turajlic, S.; Xu, H.; Litchfield, K.; Rowan, A.; Chambers, T.; Lopez, J.I.; et al. Tracking Cancer Evolution Reveals Constrained Routes to Metastases: TRACERx Renal. Cell. 2018, 173, 581–594. [Google Scholar] [CrossRef]
  130. Burrell, R.A.; Swanton, C. Tumour heterogeneity and the evolution of polyclonal drug resistance. Mol Oncol. 2014, 8, 1095–111. [Google Scholar] [CrossRef]
  131. Sottoriva, A.; Kang, H.; Ma, Z.; Graham, T.A.; Salomon, M.P.; Zhao, J.; et al. A Big Bang model of human colorectal tumor growth. Nat Genet. 2015, 47, 209–16. [Google Scholar] [CrossRef]
  132. McGranahan, N.; Swanton, C. Clonal Heterogeneity and Tumor Evolution: Past, Present, and the Future. Cell. 2017, 168, 613–28. [Google Scholar] [CrossRef]
  133. Landau, D.A.; Carter, S.L.; Stojanov, P.; McKenna, A.; Stevenson, K.; Lawrence, M.S.; et al. Evolution and Impact of Subclonal Mutations in Chronic Lymphocytic Leukemia. Cell. 2013, 152, 714–26. [Google Scholar] [CrossRef] [PubMed]
  134. Hao, J.J.; Lin, D.C.; Dinh, H.Q.; Mayakonda, A.; Jiang, Y.Y.; Chang, C.; et al. Spatial intratumoral heterogeneity and temporal clonal evolution in esophageal squamous cell carcinoma. Nat Genet. 2016, 48, 1500–7. [Google Scholar] [CrossRef] [PubMed]
  135. Lamprecht, S.; Schmidt, E.M.; Blaj, C.; Hermeking, H.; Jung, A.; Kirchner, T.; et al. Multicolor lineage tracing reveals clonal architecture and dynamics in colon cancer. Nat Commun. 2017, 8, 1406. [Google Scholar] [CrossRef] [PubMed]
  136. Larionova, I.; Tuguzbaeva, G.; Ponomaryova, A.; Stakheyeva, M.; Cherdyntseva, N.; Pavlov, V.; et al. Tumor-Associated Macrophages in Human Breast, Colorectal, Lung, Ovarian and Prostate Cancers. Front Oncol. 2020, 10, 566511. [Google Scholar] [CrossRef]
  137. Ghambashidze, K.; Chikhladze, R.; Saladze, T.; Hoopes, P.J.; Shubitidze, F.E. coli Phagelysate: A Primer to Enhance Nanoparticles and Drug Deliveries in Tumor. Cancers. 2023, 15, 2315. [Google Scholar] [CrossRef]
  138. Gambera, S.; Abarrategi, A.; González-Camacho, F.; Morales-Molina, Á.; Roma, J.; Alfranca, A.; et al. Clonal dynamics in osteosarcoma defined by RGB marking. Nat Commun. 2018, 9, 3994. [Google Scholar] [CrossRef]
  139. Semenza, G.L. The hypoxic tumor microenvironment: A driving force for breast cancer progression. Biochim Biophys Acta (BBA) - Mol Cell Res. 2016, 1863, 382–91. [Google Scholar] [CrossRef]
  140. Seth, S.; Li, C.Y.; Ho, I.L.; Corti, D.; Loponte, S.; Sapio, L.; et al. Pre-existing Functional Heterogeneity of Tumorigenic Compartment as the Origin of Chemoresistance in Pancreatic Tumors. Cell Rep. 2019, 26, 1518–1532. [Google Scholar] [CrossRef]
  141. Mustafa, S.; Jansen, C.S.; Jani, Y.; Evans, S.; Zhuang, T.Z.; Brown, J.; et al. The Evolving Landscape of Biomarkers for Immune Checkpoint Blockade in Genitourinary Cancers. Biomark Insights. 2024, 19, 11772719241254180. [Google Scholar] [CrossRef]
  142. Bravaccini, S.; Bronte, G.; Ulivi, P. TMB in NSCLC: A Broken Dream? Int J Mol Sci. 2021, 22, 6536. [Google Scholar] [CrossRef]
  143. Friedlaender, A.; Nouspikel, T.; Christinat, Y.; Ho, L.; McKee, T.; Addeo, A. Tissue-Plasma TMB Comparison and Plasma TMB Monitoring in Patients With Metastatic Non-small Cell Lung Cancer Receiving Immune Checkpoint Inhibitors. Front Oncol. 2020, 10, 142. [Google Scholar] [CrossRef] [PubMed]
  144. Elgendy, M.; Fusco, J.P.; Segura, V.; Lozano, M.D.; Minucci, S.; Echeveste, J.I.; et al. Identification of mutations associated with acquired resistance to sunitinib in renal cell cancer. Int J Cancer. 2019, 145, 1991–2001. [Google Scholar] [CrossRef] [PubMed]
  145. Jang, A.; Lanka, S.M.; Jaeger, E.B.; Lieberman, A.; Huang, M.; Sartor, A.O.; et al. Longitudinal Monitoring of Circulating Tumor DNA to Assess the Efficacy of Immune Checkpoint Inhibitors in Patients With Advanced Genitourinary Malignancies. JCO Precis Oncol. 2023, 7, e2300131. [Google Scholar] [CrossRef]
  146. Malla, M.; Loree, J.M.; Kasi, P.M.; Parikh, A.R. Using Circulating Tumor DNA in Colorectal Cancer: Current and Evolving Practices. J Clin Oncol. 2022, 40, 2846–57. [Google Scholar] [CrossRef] [PubMed]
  147. Alix-Panabières, C.; Pantel, K. Clinical Applications of Circulating Tumor Cells and Circulating Tumor DNA as Liquid Biopsy. Cancer Discov. 2016, 6, 479–91. [Google Scholar] [CrossRef]
  148. Ozdogan, M.; Papadopoulou, E.; Metaxa-Mariatou, V.; Kapetsis, G.; Meintani, A.; Florou-Chatzigiannidou, C.; et al. Case report: Immunotherapy guided by molecular profiling of tumors: illustrative cases and literature review. Front Med. 2024, 11, 1403056. [Google Scholar] [CrossRef]
  149. Krasniqi, E.; Goeman, F.; Pulito, C.; Palcau, A.C.; Ciuffreda, L.; Lisa, F.S.D.; et al. Biomarkers of Response and Resistance to CDK4/6 Inhibitors in Breast Cancer: Hints from Liquid Biopsy and microRNA Exploration. Int J Mol Sci. 2022, 23, 14534. [Google Scholar] [CrossRef]
  150. Blomain, E.S.; Moding, E.J. Liquid Biopsies for Molecular Biology-Based Radiotherapy. Int J Mol Sci. 2021, 22, 11267. [Google Scholar] [CrossRef]
  151. Siravegna, G.; Mussolin, B.; Venesio, T.; Marsoni, S.; Seoane, J.; Dive, C.; et al. How liquid biopsies can change clinical practice in oncology. Ann Oncol. 2019, 30, 1580–90. [Google Scholar] [CrossRef]
  152. Hodara, E.; Morrison, G.; Cunha, A.T.; Zainfeld, D.; Xu, T.; Xu, Y.; et al. Multi-parametric liquid biopsy analysis in metastatic prostate cancer. JCI Insight.
  153. Meo, A.D.; Bartlett, J.; Cheng, Y.; Pasic, M.D.; Yousef, G.M. Liquid biopsy: a step forward towards precision medicine in urologic malignancies. Mol Cancer. 2017, 16, 80. [Google Scholar] [CrossRef]
  154. Parisi, F.M.; Lentini, M.; Chiesa-Estomba, C.M.; Mayo-Yanez, M.; Leichen, J.R.; White, M.; et al. Liquid Biopsy in HPV-Associated Head and Neck Cancer: A Comprehensive Review. Cancers. 2025, 17, 977. [Google Scholar] [CrossRef] [PubMed]
  155. Visal, T.H.; Hollander Pden Cristofanilli, M.; Mani, S.A. Circulating tumour cells in the -omics era: how far are we from achieving the ‘singularity’? Br J Cancer. 2022, 127, 173–84. [Google Scholar] [CrossRef]
  156. Baird, B.N.; Schliekelman, M.J.; Ahn, Y.H.; Chen, Y.; Roybal, J.D.; Gill, B.J.; et al. Fibulin-2 Is a Driver of Malignant Progression in Lung Adenocarcinoma. PLoS ONE. 2013, 8, e67054. [Google Scholar] [CrossRef] [PubMed]
  157. Park, E.S.; Kim, S.J.; Kim, S.W.; Yoon, S.L.; Leem, S.H.; Kim, S.B.; et al. Cross-species hybridization of microarrays for studying tumor transcriptome of brain metastasis. Proc Natl Acad Sci. 2011, 108, 17456–61. [Google Scholar] [CrossRef]
  158. Elia, A.R.; Caputo, S.; Bellone, M. Immune Checkpoint-Mediated Interactions Between Cancer and Immune Cells in Prostate Adenocarcinoma and Melanoma. Front Immunol. 2018, 9, 1786. [Google Scholar] [CrossRef]
  159. Zhao, Y.; Weng, Z.; Zhou, X.; Xu, Z.; Cao, B.; Wang, B.; et al. Mesenchymal stromal cells promote the drug resistance of gastrointestinal stromal tumors by activating the PI3K-AKT pathway via TGF-β2. J Transl Med. 2023, 21, 219. [Google Scholar] [CrossRef] [PubMed]
  160. Straussman, R.; Morikawa, T.; Shee, K.; Barzily-Rokni, M.; Qian, Z.R.; Du, J.; et al. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature. 2012, 487, 500–4. [Google Scholar] [CrossRef]
  161. ZIF; HEJ; HED; LIY; YANGL; CAIZ Fibroblast activation protein α in tumor microenvironment: Recent progression and implications (Review). Mol Med Rep. 2015, 11, 3203–11. [CrossRef]
  162. Scioli, M.G.; Terriaca, S.; Fiorelli, E.; Storti, G.; Fabbri, G.; Cervelli, V.; et al. Extracellular Vesicles and Cancer Stem Cells in Tumor Progression: New Therapeutic Perspectives. Int J Mol Sci. 2021, 22, 10572. [Google Scholar] [CrossRef]
  163. Zhu, Y.; Knolhoff, B.L.; Meyer, M.A.; Nywening, T.M.; West, B.L.; Luo, J.; et al. CSF1/CSF1R Blockade Reprograms Tumor-Infiltrating Macrophages and Improves Response to T-cell Checkpoint Immunotherapy in Pancreatic Cancer Models. Cancer Res. 2014, 74, 5057–69. [Google Scholar] [CrossRef]
  164. Flores, C.T.; Wildes, T.J.; Drake, J.A.; Moore, G.L.; Dean, B.D.; Abraham, R.S.; et al. Lin−CCR2+ hematopoietic stem and progenitor cells overcome resistance to PD-1 blockade. Nat Commun. 2018, 9, 4313. [Google Scholar] [CrossRef] [PubMed]
  165. Hartmann, S.; Bhola, N.E.; Grandis, J.R. HGF/Met Signaling in Head and Neck Cancer: Impact on the Tumor Microenvironment. Clin Cancer Res. 2016, 22, 4005–13. [Google Scholar] [CrossRef] [PubMed]
  166. Doha, Z.O.; Sears, R.C. Unraveling MYC’s Role in Orchestrating Tumor Intrinsic and Tumor Microenvironment Interactions Driving Tumorigenesis and Drug Resistance. Pathophysiology. 2023, 30, 400–19. [Google Scholar] [CrossRef]
  167. Liang, X.; Li, Y.; Guo, B.; Zeng, Z.; Deng, K.; Zou, D.; et al. Inherent Tumor Microenvironment-Reversing Hydrogels: Potentiating Molecular Therapy Efficacy Against Drug-Resistant Tumors. Adv Funct Mater.
  168. Xing, Y.; Zhang, Y.; Li, J.; Tang, Y.; Zhang, J.; Yang, R.; et al. Bioresponsive Nanoparticles Boost Starvation Therapy and Prevent Premetastatic Niche Formation for Pulmonary Metastasis Treatment. ACS Appl Mater Interfaces. 2024, 16, 51798–806. [Google Scholar] [CrossRef]
  169. Nguyen, B.; Fong, C.; Luthra, A.; Smith, S.A.; DiNatale, R.G.; Nandakumar, S.; et al. Genomic characterization of metastatic patterns from prospective clinical sequencing of 25,000 patients. Cell. 2022, 185, 563–575. [Google Scholar] [CrossRef]
  170. Shao, X.; Lv, N.; Liao, J.; Long, J.; Xue, R.; Ai, N.; et al. Copy number variation is highly correlated with differential gene expression: a pan-cancer study. BMC Méd Genet. 2019, 20, 175. [Google Scholar] [CrossRef]
  171. Racher, H.; Soliman, S.; Argiropoulos, B.; Chan, H.S.L.; Gallie, B.L.; Perrier, R.; et al. Molecular analysis distinguishes metastatic disease from second cancers in patients with retinoblastoma. Cancer Genet.
  172. Han, Y.; Wang, C.; Dong, Q.; Chen, T.; Yang, F.; Liu, Y.; et al. Genetic Interaction-Based Biomarkers Identification for Drug Resistance and Sensitivity in Cancer Cells. Mol Ther - Nucleic Acids. 2019, 17, 688–700. [Google Scholar] [CrossRef]
  173. Martins, F.C.; Couturier, D.L.; Santiago Ide Sauer, C.M.; Vias, M.; Angelova, M.; et al. Clonal somatic copy number altered driver events inform drug sensitivity in high-grade serous ovarian cancer. Nat Commun. 2022, 13, 6360. [Google Scholar] [CrossRef]
  174. Bambury, R.M.; Bhatt, A.S.; Riester, M.; Pedamallu, C.S.; Duke, F.; Bellmunt, J.; et al. DNA copy number analysis of metastatic urothelial carcinoma with comparison to primary tumors. BMC Cancer. 2015, 15, 242. [Google Scholar] [CrossRef]
  175. Pappas, K.; Xu, J.; Zairis, S.; Resnick-Silverman, L.; Abate, F.; Steinbach, N.; et al. p53 Maintains Baseline Expression of Multiple Tumor Suppressor Genes. Mol Cancer Res. 2017, 15, 1051–62. [Google Scholar] [CrossRef]
  176. Morris, L.G.T.; Chan, T.A. Therapeutic targeting of tumor suppressor genes. Cancer. 2014, 121, 1357–68. [Google Scholar] [CrossRef] [PubMed]
  177. Davoli, T.; Xu, A.W.; Mengwasser, K.E.; Sack, L.M.; Yoon, J.C.; Park, P.J.; et al. Cumulative Haploinsufficiency and Triplosensitivity Drive Aneuploidy Patterns and Shape the Cancer Genome. Cell. 2013, 155, 948–62. [Google Scholar] [CrossRef] [PubMed]
  178. Alanee, S.; Shah, S.; Murali, R.; Rau-Murthy, R.; Schrader, K.A.; Offit, K. Absence of loss of heterozygosity of BRCA1 in a renal tumor from a BRCA1 germline mutation carrier. Fam Cancer. 2012, 12, 125–7. [Google Scholar] [CrossRef]
  179. Logothetis, C.; Morris, M.J.; Den, R.; Coleman, R.E. Current perspectives on bone metastases in castrate-resistant prostate cancer. Cancer Metastasis Rev. 2018, 37, 189–96. [Google Scholar] [CrossRef]
  180. Zhao, M.; Kim, P.; Mitra, R.; Zhao, J.; Zhao, Z. TSGene 2.0: an updated literature-based knowledgebase for tumor suppressor genes. Nucleic Acids Res. 1023. [Google Scholar]
  181. Kong, B.; Zheng, Z.; Mi, Z.; Dou, Z.; Yang, Y.; Shen, Y.; et al. Restoration of tumor suppressor protein with enhanced activity using engineered tRNAs to induce tumor regression. bioRxiv. 6432. [Google Scholar]
  182. Tomar, T.; Jong Sde Alkema, N.G.; Hoekman, R.L.; Meersma, G.J.; Klip, H.G.; et al. Genome-wide methylation profiling of ovarian cancer patient-derived xenografts treated with the demethylating agent decitabine identifies novel epigenetically regulated genes and pathways. Genome Med. 2016, 8, 107. [Google Scholar] [CrossRef]
  183. Matthews, B.G.; Bowden, N.A.; Wong-Brown, M.W. Epigenetic Mechanisms and Therapeutic Targets in Chemoresistant High-Grade Serous Ovarian Cancer. Cancers. 2021, 13, 5993. [Google Scholar] [CrossRef]
  184. Bai, Z.T.; Bai, B.; Zhu, J.; Di, C.X.; Li, X.; Zhou, W.C. Epigenetic actions of environmental factors and promising drugs for cancer therapy. Oncol Lett. 2017, 15, 2049–56. [Google Scholar] [CrossRef]
  185. Castro-Muñoz, L.J.; Ulloa, E.V.; Sahlgren, C.; Lizano, M.; Cruz-Hernández, E.D.L.; Contreras-Paredes, A. Modulating epigenetic modifications for cancer therapy (Review). Oncol Rep. 2023, 49, 59. [Google Scholar] [CrossRef]
  186. Bennett, R.L.; Licht, J.D. Targeting Epigenetics in Cancer. Annu Rev Pharmacol Toxicol. 2018, 58, 1–21. [Google Scholar] [CrossRef]
  187. Toh, T.B.; Lim, J.J.; Chow, E.K.H. Epigenetics in cancer stem cells. Mol Cancer. 2017, 16, 29. [Google Scholar] [CrossRef]
  188. Chen, C.; Guo, Y.; Guo, Y.; Wu, X.; Si, C.; Xu, Y.; et al. m6A Modification in Non-Coding RNA: The Role in Cancer Drug Resistance. Front Oncol. 2021, 11, 746789. [Google Scholar] [CrossRef] [PubMed]
  189. Panda, R.; Mohan, S.; Vellapandian, C. Harnessing Epigenetic Mechanisms to Overcome Immune Evasion in Cancer: The Current Strategies and Future Directions. Cureus. 2024, 16, e70631. [Google Scholar] [CrossRef] [PubMed]
  190. Rubatto, M.; Borriello, S.; Sciamarrelli, N.; Pala, V.; Tonella, L.; Ribero, S.; et al. Exploring the role of epigenetic alterations and non-coding RNAs in melanoma pathogenesis and therapeutic strategies. Melanoma Res. 2023, 33, 462–74. [Google Scholar] [CrossRef] [PubMed]
  191. Liau, B.B.; Sievers, C.; Donohue, L.K.; Gillespie, S.M.; Flavahan, W.A.; Miller, T.E.; et al. Adaptive Chromatin Remodeling Drives Glioblastoma Stem Cell Plasticity and Drug Tolerance. Cell Stem Cell. 2017, 20, 233–246. [Google Scholar] [CrossRef]
  192. Bao, Y.; Oguz, G.; Lee, W.C.; Lee, P.L.; Ghosh, K.; Li, J.; et al. EZH2-mediated PP2A inactivation confers resistance to HER2-targeted breast cancer therapy. Nat Commun. 2020, 11, 5878. [Google Scholar] [CrossRef]
  193. Anderson, N.M.; Simon, M.C. The tumor microenvironment. Curr Biol. 2020, 30, R921–5. [Google Scholar] [CrossRef]
  194. Somasundaram, R.; Herlyn, M.; Wagner, S.N. The role of tumor microenvironment in melanoma therapy resistance. Melanoma Manag. 2016, 3, 23–32. [Google Scholar] [CrossRef]
  195. Zhang, Q.; Yang, J.; Bai, J.; Ren, J. Reverse of non-small cell lung cancer drug resistance induced by cancer-associated fibroblasts via a paracrine pathway. Cancer Sci. 2018, 109, 944–55. [Google Scholar] [CrossRef]
  196. Wang, Z.; Liu, J.; Huang, H.; Ye, M.; Li, X.; Wu, R.; et al. Metastasis-associated fibroblasts: an emerging target for metastatic cancer. Biomark Res. 2021, 9, 47. [Google Scholar] [CrossRef]
  197. Behera, R.; Kaur, A.; Webster, M.R.; Kim, S.; Ndoye, A.; Kugel, C.H.; et al. Inhibition of Age-Related Therapy Resistance in Melanoma by Rosiglitazone-Mediated Induction of Klotho. Clin Cancer Res. 2017, 23, 3181–90. [Google Scholar] [CrossRef]
  198. Domukhovska, A.; Burakgazi, Z.A.; Springer, M.; Nafchi, B.; Beary, M.C.; Acquisto, A.; et al. Hif-1a-Mediated Disruption of Cellular Junctions: The Impact of Hypoxia on the Tumor Microenvironment and Invasion. 2025. [Google Scholar]
  199. Lv, X.; Li, J.; Zhang, C.; Hu, T.; Li, S.; He, S.; et al. The role of hypoxia-inducible factors in tumor angiogenesis and cell metabolism. Genes Dis. 2017, 4, 19–24. [Google Scholar] [CrossRef] [PubMed]
  200. Kim, J.Y.; Lee, J.Y. Targeting Tumor Adaption to Chronic Hypoxia: Implications for Drug Resistance, and How It Can Be Overcome. Int J Mol Sci. 2017, 18, 1854. [Google Scholar] [CrossRef]
  201. Kumar, H.; Choi, D.K. Hypoxia Inducible Factor Pathway and Physiological Adaptation: A Cell Survival Pathway? Mediat Inflamm. 2015, 2015, 584758. [Google Scholar] [CrossRef]
  202. Zheng, X.; Fan, H.; Liu, Y.; Wei, Z.; Li, X.; Wang, A.; et al. Hypoxia Boosts Aerobic Glycolysis in Carcinoma: A Complex Process for Tumour Development. Curr Mol Pharmacol. 2022, 15, 487–501. [Google Scholar] [CrossRef] [PubMed]
  203. Chipurupalli, S.; Kannan, E.; Tergaonkar, V.; D’Andrea, R.; Robinson, N. Hypoxia Induced ER Stress Response as an Adaptive Mechanism in Cancer. Int J Mol Sci. 2019, 20, 749. [Google Scholar] [CrossRef] [PubMed]
  204. Leiphrakpam, P.D.; Are, C. PI3K/Akt/mTOR Signaling Pathway as a Target for Colorectal Cancer Treatment. Int J Mol Sci. 2024, 25, 3178. [Google Scholar] [CrossRef]
  205. Pungsrinont, T.; Kallenbach, J.; Baniahmad, A. Role of PI3K-AKT-mTOR Pathway as a Pro-Survival Signaling and Resistance-Mediating Mechanism to Therapy of Prostate Cancer. Int J Mol Sci. 2021, 22, 11088. [Google Scholar] [CrossRef]
  206. Dong, C.; Wu, J.; Chen, Y.; Nie, J.; Chen, C. Activation of PI3K/AKT/mTOR Pathway Causes Drug Resistance in Breast Cancer. Front Pharmacol. 2021, 12, 628690. [Google Scholar] [CrossRef]
  207. Yang, J.; Nie, J.; Ma, X.; Wei, Y.; Peng, Y.; Wei, X. Targeting PI3K in cancer: mechanisms and advances in clinical trials. Mol Cancer. 2019, 18, 26. [Google Scholar] [CrossRef]
  208. Schwartz, S.; Wongvipat, J.; Trigwell, C.B.; Hancox, U.; Carver, B.S.; Rodrik-Outmezguine, V.; et al. Feedback Suppression of PI3Kα Signaling in PTEN-Mutated Tumors Is Relieved by Selective Inhibition of PI3Kβ. Cancer Cell. 2015, 27, 109–22. [Google Scholar] [CrossRef]
  209. Amin, T.; Viol, F.; Krause, J.; Fahl, M.; Eggers, C.; Awwad, F.; et al. Cancer-Associated Fibroblasts Induce Proliferation and Therapeutic Resistance to Everolimus in Neuroendocrine Tumors through STAT3 Activation. Neuroendocrinology. 2022, 113, 501–18. [Google Scholar] [CrossRef] [PubMed]
  210. Thakur, N.; Singh, P.; Bagri, A.; Srivastava, S.; Dwivedi, V.; Singh, A.; et al. Therapy resistance in prostate cancer: mechanism, signaling and reversal strategies. Explor Target Anti-tumor Ther. 2024, 5, 1110–34. [Google Scholar] [CrossRef] [PubMed]
  211. Crowley, F.; Sterpi, M.; Buckley, C.; Margetich, L.; Handa, S.; Dovey, Z. A Review of the Pathophysiological Mechanisms Underlying Castration-resistant Prostate Cancer. Res Rep Urol. 2021, 13, 457–72. [Google Scholar] [CrossRef]
  212. Tran, M.G.B.; Bibby, B.A.S.; Yang, L.; Lo, F.; Warren, A.Y.; Shukla, D.; et al. Independence of HIF1a and androgen signaling pathways in prostate cancer. BMC Cancer. 2020, 20, 469. [Google Scholar] [CrossRef] [PubMed]
  213. Fernandez, E.V.; Reece, K.M.; Ley, A.M.; Troutman, S.M.; Sissung, T.M.; Price, D.K.; et al. Dual Targeting of the Androgen Receptor and Hypoxia-Inducible Factor 1α Pathways Synergistically Inhibits Castration-Resistant Prostate Cancer Cells. Mol Pharmacol. 2015, 87, 1006–12. [Google Scholar] [CrossRef]
  214. Lee, A.C.K.; Lau, P.M.; Kwan, Y.W.; Kong, S.K. Mitochondrial Fuel Dependence on Glutamine Drives Chemo-Resistance in the Cancer Stem Cells of Hepatocellular Carcinoma. Int J Mol Sci. 2021, 22, 3315. [Google Scholar] [CrossRef]
  215. Crispim, D.; Ramos, C.; Esteves, F.; Kranendonk, M. The Adaptation of MCF-7 Breast Cancer Spheroids to the Chemotherapeutic Doxorubicin: The Dynamic Role of Phase I Drug Metabolizing Enzymes. Metabolites. 2025, 15, 136. [Google Scholar] [CrossRef] [PubMed]
  216. Lin, J.; Xia, L.; Oyang, L.; Liang, J.; Tan, S.; Wu, N.; et al. The POU2F1-ALDOA axis promotes the proliferation and chemoresistance of colon cancer cells by enhancing glycolysis and the pentose phosphate pathway activity. Oncogene. 2022, 41, 1024–39. [Google Scholar] [CrossRef] [PubMed]
  217. Lv, L.; Yang, S.; Zhu, Y.; Zhai, X.; Li, S.; Tao, X.; et al. Relationship between metabolic reprogramming and drug resistance in breast cancer. Front Oncol. 2022, 12, 942064. [Google Scholar] [CrossRef]
  218. McCann, C.; Kerr, E.M. Metabolic Reprogramming: A Friend or Foe to Cancer Therapy? Cancers. 2021, 13, 3351. [Google Scholar] [CrossRef]
  219. Germain, N.; Dhayer, M.; Boileau, M.; Fovez, Q.; Kluza, J.; Marchetti, P. Lipid Metabolism and Resistance to Anticancer Treatment. Biology. 2020, 9, 474. [Google Scholar] [CrossRef] [PubMed]
  220. Criscuolo, D.; Avolio, R.; Calice, G.; Laezza, C.; Paladino, S.; Navarra, G.; et al. Cholesterol Homeostasis Modulates Platinum Sensitivity in Human Ovarian Cancer. Cells. 2020, 9, 828. [Google Scholar] [CrossRef]
  221. Ponton-Almodovar, A.; Udumula, M.P.; Khullar, V.; Rashid, F.; Rattan, R.; Bernard, J.J.; et al. GPT2 mediates metabolic alterations in platinum-resistant ovarian cancer cells. Res Sq. 6480. [Google Scholar]
  222. Bort, A.; Sánchez, B.G.; León, C.; Nozal, L.; Mora-Rodríguez, J.M.; Castro, F.; et al. Metabolic fingerprinting of chemotherapy-resistant prostate cancer stem cells. An untargeted metabolomic approach by liquid chromatography-mass spectrometry. Front Cell Dev Biol. 2022, 10, 1005675. [Google Scholar] [CrossRef]
  223. Shi, J.; Shen, Y.; Zhang, J. Emerging roles of small extracellular vesicles in metabolic reprogramming and drug resistance in cancers. Cancer Drug Resist. 2024, 7, N/A–N/A. [Google Scholar] [CrossRef] [PubMed]
  224. Kou, Z.; Liu, C.; Zhang, W.; Sun, C.; Liu, L.; Zhang, Q. Heterogeneity of primary and metastatic CAFs: From differential treatment outcomes to treatment opportunities (Review). Int J Oncol. 2024, 64, 54. [Google Scholar] [CrossRef]
  225. Zhang, L.; Zhang, S.; Yao, J.; Lowery, F.J.; Zhang, Q.; Huang, W.C.; et al. Microenvironment-induced PTEN loss by exosomal microRNA primes brain metastasis outgrowth. Nature. 2015, 527, 100–4. [Google Scholar] [CrossRef] [PubMed]
  226. Lee, C.C.; Lin, J.C.; Hwang, W.L.; Kuo, Y.J.; Chen, H.K.; Tai, S.K.; et al. Macrophage-secreted interleukin-35 regulates cancer cell plasticity to facilitate metastatic colonization. Nat Commun. 2018, 9, 3763. [Google Scholar] [CrossRef]
  227. Michaud, D.E.; Guerriero, J.L. Myeloid Cells Pave the Metastatic Road in Breast Cancer. Cancer Res. 2023, 84, 181–3. [Google Scholar] [CrossRef]
  228. Gouirand, V.; Guillaumond, F.; Vasseur, S. Influence of the Tumor Microenvironment on Cancer Cells Metabolic Reprogramming. Front Oncol. 2018, 8, 117. [Google Scholar] [CrossRef]
  229. Lehuédé, C.; Dupuy, F.; Rabinovitch, R.; Jones, R.G.; Siegel, P.M. Metabolic Plasticity as a Determinant of Tumor Growth and Metastasis. Cancer Res. 2016, 76, 5201–8. [Google Scholar] [CrossRef]
  230. Rushing, B.R.; Molina, S.; Sumner, S. Metabolomics Analysis Reveals Altered Metabolic Pathways and Response to Doxorubicin in Drug-Resistant Triple-Negative Breast Cancer Cells. Metabolites. 2023, 13, 865. [Google Scholar] [CrossRef] [PubMed]
  231. Rozenblit, M.; Huang, R.; Danziger, N.; Hegde, P.; Alexander, B.; Ramkissoon, S.; et al. Comparison of PD-L1 protein expression between primary tumors and metastatic lesions in triple negative breast cancers. J Immunother Cancer. 2020, 8, e001558. [Google Scholar] [CrossRef] [PubMed]
  232. Xu, B.; Chen, J.; Sarungbam, J.; Tickoo, S.; Dickson, B.C.; Reuter, V.E.; et al. NUTM1-fusion positive malignant neoplasms of the genitourinary tract: A report of six cases highlighting involvement of unusual anatomic locations and histologic heterogeneity. Genes, Chromosom Cancer. 2022, 61, 542–50. [Google Scholar] [CrossRef]
  233. Luo, W.; Stevens, T.M.; Stafford, P.; Miettinen, M.; Gatalica, Z.; Vranic, S. NUTM1-Rearranged Neoplasms: A Heterogeneous Group of Primitive Tumors with Expanding Spectrum of Histology and Molecular Alterations: An Updated Review. Preprints. 2109. [Google Scholar]
  234. McEvoy, C.R.; Fox, S.B.; Prall, O.W.J. Emerging entities in NUTM1-rearranged neoplasms. Genes, Chromosom Cancer. 2020, 59, 375–85. [Google Scholar] [CrossRef]
  235. Abida, W.; Cheng, M.L.; Armenia, J.; Middha, S.; Autio, K.A.; Vargas, H.A.; et al. Analysis of the Prevalence of Microsatellite Instability in Prostate Cancer and Response to Immune Checkpoint Blockade. JAMA Oncol. 2019, 5, 471–8. [Google Scholar] [CrossRef]
  236. Ukleja, J.; Kusaka, E.; Miyamoto, D.T. Immunotherapy Combined With Radiation Therapy for Genitourinary Malignancies. Front Oncol. 2021, 11, 663852. [Google Scholar] [CrossRef]
  237. Herr, H.; Sogani, P.; Eastham, J. Genitourinary tumors. J Surg Oncol. 2022, 126, 926–32. [Google Scholar] [CrossRef] [PubMed]
  238. Zhou, Z.; Cui, Y.; Zhang, Y.; Wu, J. Editorial: The functional role of non-coding RNAs in tumor microenvironment and metastasis of genitourinary tumor and its potential application as tumor molecular biomarkers. Front Genet. 2023, 14, 1133496. [Google Scholar] [CrossRef]
  239. Gareev, I.; Gileva, Y.; Dzidzaria, A.; Beylerli, O.; Pavlov, V.; Agaverdiev, M.; et al. Long non-coding RNAs in oncourology. Non-coding RNA Res. 2021, 6, 139–45. [Google Scholar] [CrossRef]
  240. Wang, R.; Du, N.; Jin, L.; Chen, W.; Ma, Z.; Zhang, T.; et al. Hyaluronic Acid Modified Au@SiO2@Au Nanoparticles for Photothermal Therapy of Genitourinary Tumors. Polymers. 2022, 14, 4772. [Google Scholar] [CrossRef]
  241. Yanovsky, R.L.; Bartenstein, D.W.; Rogers, G.S.; Isakoff, S.J.; Chen, S.T. Photodynamic therapy for solid tumors: A review of the literature. Photodermatol, Photoimmunol Photomed. 2019, 35, 295–303. [Google Scholar] [CrossRef] [PubMed]
  242. Nathan, P.; Rajeh, A.; Noor, M.; Boldt, G.; Fernandes, R. Antibody–Drug Conjugates in the Treatment of Genitourinary Cancers: An Updated Review of Data. Curr Oncol. 2024, 31, 2316–27. [Google Scholar] [CrossRef] [PubMed]
  243. Martin, S.K.; Pu, H.; Penticuff, J.C.; Cao, Z.; Horbinski, C.; Kyprianou, N. Multinucleation and Mesenchymal-to-Epithelial Transition Alleviate Resistance to Combined Cabazitaxel and Antiandrogen Therapy in Advanced Prostate Cancer. Cancer Res. 2016, 76, 912–26. [Google Scholar] [CrossRef]
  244. Wattenberg, M.M.; Fong, L.; Madan, R.A.; Gulley, J.L. Immunotherapy in genitourinary malignancies. Curr Opin Urol. 2016, 26, 501–7. [Google Scholar] [CrossRef] [PubMed]
  245. Obinata, D.; Hashimoto, S.; Uchida, H.; Nakahara, K.; Yoshizawa, T.; Mochida, J.; et al. Clinical characteristics of patients with metastatic castration-resistant prostate cancer after treatment with combined androgen blockade. BMC Urol. 2023, 23, 74. [Google Scholar] [CrossRef]
  246. McKay, R.R.; Bossé, D.; Choueiri, T.K. Evolving Systemic Treatment Landscape for Patients With Advanced Renal Cell Carcinoma. J Clin Oncol. 2018, 36, 3615–23. [Google Scholar] [CrossRef]
  247. Weinstock, M.; McDermott, D. Targeting PD-1/PD-L1 in the treatment of metastatic renal cell carcinoma. Ther Adv Urol. 2015, 7, 365–77. [Google Scholar] [CrossRef]
  248. Costanzo, F.D.; Napolitano, F.; Salomone, F.; Amato, A.R.; Alberico, G.; Migliaccio, F.; et al. Analysis of Health-Related Quality of Life Reporting in Phase III RCTs of Advanced Genitourinary Tumors. Cancers. 2023, 15, 5703. [Google Scholar] [CrossRef]
  249. Malik, A.; Srinivasan, S.; Batra, J. A New Era of Prostate Cancer Precision Medicine. Front Oncol. 2019, 9, 1263. [Google Scholar] [CrossRef]
  250. Kamran, S.C.; Efstathiou, J.A. Current State of Personalized Genitourinary Cancer Radiotherapy in the Era of Precision Medicine. Front Oncol. 2021, 11, 675311. [Google Scholar] [CrossRef]
  251. Takeuchi, S.; Yoshimura, A.; Sofuni, A.; Ueda, Y.; Umezu, T.; Kuroda, M.; et al. A single-institution retrospective study of comprehensive genomic profiling tests based on C-CAT findings for advanced solid cancers. Jpn J Clin Oncol. 2024, 54, 1298–305. [Google Scholar] [CrossRef] [PubMed]
  252. Matsubara, N.; Kato, T.; Fujisawa, T.; Shiota, M.; Eto, M.; Osawa, T.; et al. Landscape of genomic alterations of circulating tumor DNA in advanced genitourinary cancer patients: SCRUM-Japan MONSTAR SCREEN Project. J Clin Oncol.
  253. Jia, E.; Zheng, T.; Huang, Y.; Du, P. Exome-wide molecular insights from blood and urine liquid biopsies in genitourinary cancers. UroPrecision. 2025. [CrossRef]
  254. Jiang, J.; Yan, Y.; Luo, Z.; He, J.; Yu, T.; Du, W.; et al. Abstract 3982: Prediction and selection of cancer drug treatments using personalized tumor models or models with matching genomic profiles. Cancer Res. 3982. [Google Scholar]
  255. Gao, S.; Soares, F.; Wang, S.; Wong, C.C.; Chen, H.; Yang, Z.; et al. CRISPR screens identify cholesterol biosynthesis as a therapeutic target on stemness and drug resistance of colon cancer. Oncogene. 2021, 40, 6601–13. [Google Scholar] [CrossRef]
  256. Jüptner, M.; Marx, M.; Zuhayra, M.; Lützen, U. Experimental 177Lu-PSMA-617 radioligand therapy in a patient with extended metastasized leiomyosarcoma. Nuklearmedizin. 2019, 58, 328–30. [Google Scholar] [CrossRef]
  257. Lee, M.; Hirpara, J.L.; Eu, J.Q.; Sethi, G.; Wang, L.; Goh, B.C.; et al. Targeting STAT3 and oxidative phosphorylation in oncogene-addicted tumors. Redox Biol. 2019, 25, 101073. [Google Scholar] [CrossRef]
  258. Alexidis, P.; Dragoumis, D.; Karatzoglou, S.; Drevelegas, K.; Tzitzikas, I.; Hatzimouratidis, K.; et al. The role of hypofractionated radiotherapy for the definitive treatment of localized prostate cancer: early results of a randomized trial. J Cancer. 2019, 10, 6217–24. [Google Scholar] [CrossRef]
  259. Ye, Y.; Hu, Q.; Chen, H.; Liang, K.; Yuan, Y.; Xiang, Y.; et al. Characterization of hypoxia-associated molecular features to aid hypoxia-targeted therapy. Nat Metab. 2019, 1, 431–44. [Google Scholar] [CrossRef] [PubMed]
  260. Nunes, T.; Hamdan, D.; Leboeuf, C.; Bouchtaoui, M.E.; Gapihan, G.; Nguyen, T.T.; et al. Targeting Cancer Stem Cells to Overcome Chemoresistance. Int J Mol Sci. 2018, 19, 4036. [Google Scholar] [CrossRef]
  261. Nguyen, L.L.; Watson, Z.L.; Ortega, R.; Woodruff, E.R.; Jordan, K.R.; Iwanaga, R.; et al. Combinatory EHMT and PARP inhibition induces an interferon response and a CD8 T cell-dependent tumor regression in PARP inhibitor-resistant models. bioRxiv. 5297. [Google Scholar]
  262. Xiao, M.; Benoit, A.; Hasmim, M.; Duhem, C.; Vogin, G.; Berchem, G.; et al. Targeting Cytoprotective Autophagy to Enhance Anticancer Therapies. Front Oncol. 2021, 11, 626309. [Google Scholar] [CrossRef]
  263. Steen, N.V.D.; Giovannetti, E.; Carbone, D.; Leonetti, A.; Rolfo, C.D.; Peters, G.J. Resistance to epidermal growth factor receptor inhibition in non-small cell lung cancer. Cancer Drug Resist. 2018, 1, 230–49. [Google Scholar]
  264. Jiang, Z.; Gu, Z.; Yu, X.; Cheng, T.; Liu, B. Research progress on the role of bypass activation mechanisms in resistance to tyrosine kinase inhibitors in non-small cell lung cancer. Front Oncol. 2024, 14, 1447678. [Google Scholar] [CrossRef] [PubMed]
  265. Huang, Q.; Yang, J.; Liu, G.X.; Zi, H.; Tang, S.D.; Jia, H.C.; et al. Changes in disease burden and global inequalities in bladder, kidney and prostate cancers from 1990 to 2019: a comparative analysis based on the global burden of disease study 2019. BMC Public Heal. 2024, 24, 891. [Google Scholar] [CrossRef]
  266. Arora, V.K.; Schenkein, E.; Murali, R.; Subudhi, S.K.; Wongvipat, J.; Balbas, M.D.; et al. Glucocorticoid Receptor Confers Resistance to Antiandrogens by Bypassing Androgen Receptor Blockade. Cell. 2013, 155, 1309–22. [Google Scholar] [CrossRef] [PubMed]
  267. Chen, R.; Dong, X.; Gleave, M. Molecular model for neuroendocrine prostate cancer progression. BJU Int. 2018, 122, 560–70. [Google Scholar] [CrossRef]
  268. Puhr, M.; Hoefer, J.; Eigentler, A.; Ploner, C.; Handle, F.; Schaefer, G.; et al. The Glucocorticoid Receptor Is a Key Player for Prostate Cancer Cell Survival and a Target for Improved Antiandrogen Therapy. Clin Cancer Res. 2018, 24, 927–38. [Google Scholar] [CrossRef]
  269. Voortman, J.; Chęcińska, A.; Giaccone, G. The proteasomal and apoptotic phenotype determine bortezomib sensitivity of non-small cell lung cancer cells. Mol Cancer. 2007, 6, 73. [Google Scholar] [CrossRef]
  270. Bluemn, E.G.; Coleman, I.M.; Lucas, J.M.; Coleman, R.T.; Hernandez-Lopez, S.; Tharakan, R.; et al. Androgen Receptor Pathway-Independent Prostate Cancer Is Sustained through FGF Signaling. Cancer Cell. 2017, 32, 474–489. [Google Scholar] [CrossRef]
  271. Wheeler, S.E.; Shi, H.; Lin, F.; Dasari, S.; Bednash, J.; Thorne, S.; et al. Enhancement of head and neck squamous cell carcinoma proliferation, invasion, and metastasis by tumor-associated fibroblasts in preclinical models. Head Neck. 2014, 36, 385–92. [Google Scholar] [CrossRef] [PubMed]
  272. Plava, J.; Cihova, M.; Burikova, M.; Matuskova, M.; Kucerova, L.; Miklikova, S. Recent advances in understanding tumor stroma-mediated chemoresistance in breast cancer. Mol Cancer. 2019, 18, 67. [Google Scholar] [CrossRef]
  273. Ruan, K.; Song, G.; Ouyang, G. Role of hypoxia in the hallmarks of human cancer. J Cell Biochem. 2009, 107, 1053–62. [Google Scholar] [CrossRef]
  274. Li, Y.; Zhang, J.; Xu, J.; Liu, S. The Metabolism Symbiosis Between Pancreatic Cancer and Tumor Microenvironment. Front Oncol. 2021, 11, 759376. [Google Scholar] [CrossRef] [PubMed]
  275. Yang, X.; Zhang, X.; Fu, M.L.; Weichselbaum, R.R.; Gajewski, T.F.; Guo, Y.; et al. Targeting the Tumor Microenvironment with Interferon-β Bridges Innate and Adaptive Immune Responses. Cancer Cell. 2014, 25, 37–48. [Google Scholar] [CrossRef] [PubMed]
  276. Guo, Z.; Jiang, Y.; Ou, B.; Lu, X.; Cheng, X.; Zhao, R. Editorial: The role of angiogenesis and immune response in tumor microenvironment of solid tumor. Front Immunol. 2023, 14, 1195390. [Google Scholar] [CrossRef]
  277. Baghban, R.; Roshangar, L.; Jahanban-Esfahlan, R.; Seidi, K.; Ebrahimi-Kalan, A.; Jaymand, M.; et al. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun Signal. 2020, 18, 59. [Google Scholar] [CrossRef]
  278. Kalli, M.; Li, R.; Mills, G.B.; Stylianopoulos, T.; Zervantonakis, I.K. Mechanical stress in pancreatic cancer: Signaling pathway adaptation activates cytoskeletal remodeling and enhances cell migration. bioRxiv. 4480. [Google Scholar]
  279. Schmid, M.C.; Varner, J.A. Myeloid Cells in the Tumor Microenvironment: Modulation of Tumor Angiogenesis and Tumor Inflammation. J Oncol. 2010, 2010, 201026. [Google Scholar] [CrossRef]
  280. Li, J.L.; Sainson, R.C.A.; Oon, C.E.; Turley, H.; Leek, R.; Sheldon, H.; et al. DLL4-Notch Signaling Mediates Tumor Resistance to Anti-VEGF Therapy In Vivo. Cancer Res. 2011, 71, 6073–83. [Google Scholar] [CrossRef]
  281. Klampfer, L. Cytokines, Inflammation and Colon Cancer. Curr Cancer Drug Targets. 2011, 11, 451–64. [Google Scholar] [CrossRef]
  282. Salemme, V.; Centonze, G.; Avalle, L.; Natalini, D.; Piccolantonio, A.; Arina, P.; et al. The role of tumor microenvironment in drug resistance: emerging technologies to unravel breast cancer heterogeneity. Front Oncol. 2023, 13, 1170264. [Google Scholar] [CrossRef]
  283. Xiang, Y.; Liu, X.; Wang, Y.; Zheng, D.; Meng, Q.; Jiang, L.; et al. Mechanisms of resistance to targeted therapy and immunotherapy in non-small cell lung cancer: promising strategies to overcoming challenges. Front Immunol. 2024, 15, 1366260. [Google Scholar] [CrossRef]
  284. Sant, M.; Bernat-Peguera, A.; Felip, E.; Margelí, M. Role of ctDNA in Breast Cancer. Cancers. 2022, 14, 310. [Google Scholar] [CrossRef]
  285. Yang, Y.; Liu, H.; Chen, Y.; Xiao, N.; Zheng, Z.; Liu, H.; et al. Liquid biopsy on the horizon in immunotherapy of non-small cell lung cancer: current status, challenges, and perspectives. Cell Death Dis. 2023, 14, 230. [Google Scholar] [CrossRef] [PubMed]
  286. Main, S.C.; Cescon, D.W.; Bratman, S.V. Liquid biopsies to predict CDK4/6 inhibitor efficacy and resistance in breast cancer. Cancer Drug Resist. 2022, 5, 727–48. [Google Scholar] [CrossRef]
  287. Goodall, J.; investigators for the, T.A.; Mateo, J.; Yuan, W.; Mossop, H.; Porta, N.; et al. Circulating Cell-Free DNA to Guide Prostate Cancer Treatment with PARP Inhibition. Cancer Discov. 2017, 7, 1006–17. [Google Scholar] [CrossRef]
  288. Battaglin, F.; Lenz, H.J. Clinical Applications of Circulating Tumor DNA Profiling in GI Cancers. JCO Oncol Pr. 2024, 20, 1481–90. [Google Scholar] [CrossRef]
  289. Thompson, J.C.; Yee, S.S.; Troxel, A.B.; Savitch, S.L.; Fan, R.; Balli, D.; et al. Detection of Therapeutically Targetable Driver and Resistance Mutations in Lung Cancer Patients by Next-Generation Sequencing of Cell-Free Circulating Tumor DNA. Clin Cancer Res. 2016, 22, 5772–82. [Google Scholar] [CrossRef]
  290. Jiang, M.; Jin, S.; Han, J.; Li, T.; Shi, J.; Zhong, Q.; et al. Detection and clinical significance of circulating tumor cells in colorectal cancer. Biomark Res. 2021, 9, 85. [Google Scholar] [CrossRef] [PubMed]
  291. Lianidou, E.; Pantel, K. Liquid biopsies. Genes, Chromosom Cancer. 2019, 58, 219–32. [Google Scholar] [CrossRef] [PubMed]
  292. Alemzadeh, E.; Allahqoli, L.; Dehghan, H.; Mazidimoradi, A.; Ghasempour, A.; Salehiniya, H. Circulating tumor cells and circulating tumor DNA in breast cancer diagnosis and monitoring. Oncol Res. 2023, 31, 667–75. [Google Scholar] [CrossRef]
  293. McDonald, B.R.; Contente-Cuomo, T.; Sammut, S.J.; Odenheimer-Bergman, A.; Ernst, B.; Perdigones, N.; et al. Personalized circulating tumor DNA analysis to detect residual disease after neoadjuvant therapy in breast cancer. Sci Transl Med.
  294. Fraipont Fde Gazzeri, S.; Cho, W.C.; Eymin, B. Circular RNAs and RNA Splice Variants as Biomarkers for Prognosis and Therapeutic Response in the Liquid Biopsies of Lung Cancer Patients. Front Genet. 2019, 10, 390. [Google Scholar] [CrossRef]
  295. Gailhouste, L.; Liew, L.C.; Hatada, I.; Nakagama, H.; Ochiya, T. Epigenetic reprogramming using 5-azacytidine promotes an anti-cancer response in pancreatic adenocarcinoma cells. Cell Death Dis. 2018, 9, 468. [Google Scholar] [CrossRef]
  296. Sun, F.; Li, L.; Yan, P.; Zhou, J.; Shapiro, S.D.; Xiao, G.; et al. Causative role of PDLIM2 epigenetic repression in lung cancer and therapeutic resistance. Nat Commun. 2019, 10, 5324. [Google Scholar] [CrossRef]
  297. Griffiths, C.; Bilbao, M.; Krill, L.; Ostrovsky, O. Ovarian Cancer - Updates in Tumour Biology and Therapeutics [Working Title]. 2021. [Google Scholar]
  298. Ari, F.; Napieralski, R.; Akgun, O.; Magdolen, V.; Ulukaya, E. Epigenetic modulators combination with chemotherapy in breast cancer cells. Cell Biochem Funct. 2021, 39, 571–83. [Google Scholar] [CrossRef] [PubMed]
  299. Feng, S.; Carvalho, D.D.D. Clinical advances in targeting epigenetics for cancer therapy. FEBS J. 2022, 289, 1214–39. [Google Scholar] [CrossRef]
  300. Liu, Z.; Ren, Y.; Weng, S.; Xu, H.; Li, L.; Han, X. A New Trend in Cancer Treatment: The Combination of Epigenetics and Immunotherapy. Front Immunol. 2022, 13, 809761. [Google Scholar] [CrossRef] [PubMed]
  301. Dominici, C.; Sgarioto, N.; Yu, Z.; Sesma-Sanz, L.; Masson, J.Y.; Richard, S.; et al. Synergistic effects of type I PRMT and PARP inhibitors against non-small cell lung cancer cells. Clin Epigenetics. 2021, 13, 54. [Google Scholar] [CrossRef] [PubMed]
  302. Shi, J.; Chen, Y.; Peng, C.; Kuang, L.; Zhang, Z.; Li, Y.; et al. Advances in Targeted Therapy Against Driver Mutations and Epigenetic Alterations in Non-Small Cell Lung Cancer. Oncologie. 2022, 24, 613–48. [Google Scholar] [CrossRef]
  303. Sookram, J.; Zheng, A.; Linden, K.M.; Morgan, A.B.; Brown, S.A.; Ostrovsky, O. Epigenetic therapy can inhibit growth of ovarian cancer cells and reverse chemoresistant properties acquired from metastatic omentum. Int J Gynecol Obstet. 2019, 145, 225–32. [Google Scholar] [CrossRef]
  304. Rimar, K.J.; Tran, P.T.; Matulewicz, R.S.; Hussain, M.; Meeks, J.J. The emerging role of homologous recombination repair and PARP inhibitors in genitourinary malignancies. Cancer. 2017, 123, 1912–24. [Google Scholar] [CrossRef]
  305. Palliyage, G.H.; Ghosh, R.; Rojanasakul, Y. Cancer chemoresistance and therapeutic strategies targeting tumor microenvironment. ScienceAsia. 2020, 46, 639. [Google Scholar] [CrossRef]
  306. Gupta, S.; Smith, T.R.; Broekman, M.L.D. Ethics of Innovation in Neurosurgery. 2019;121–8.
  307. Sindhu, K.K.; Dovey, Z.; Thompson, M.; Nehlsen, A.D.; Skalina, K.A.; Malachowska, B.; et al. The potential role of precision medicine to alleviate racial disparities in prostate, bladder and renal urological cancer care. BJUI Compass. 2024, 5, 405–25. [Google Scholar] [CrossRef]
  308. Vo, H.H.; Fu, S.; Hong, D.S.; Karp, D.D.; Piha-Paul, S.; Subbiah, V.; et al. Challenges and opportunities associated with the MD Anderson IMPACT2 randomized study in precision oncology. npj Precis Oncol. 2022, 6, 78. [Google Scholar] [CrossRef] [PubMed]
  309. Song, K.W.; Wen, P.Y. Novel trial designs in neuro-oncology. Curr Opin Neurol. 2023, 36, 571–8. [Google Scholar] [CrossRef]
  310. McClatchy, D.M.; Willers, H.; Hata, A.N.; Piotrowska, Z.; Sequist, L.V.; Paganetti, H.; et al. Modeling Resistance and Recurrence Patterns of Combined Targeted–Chemoradiotherapy Predicts Benefit of Shorter Induction Period. Cancer Res. 2020, 80, 5121–33. [Google Scholar] [CrossRef]
  311. Finzel, A.; Sadik, H.; Ghitti, G.; Laes, J.F. The combined analysis of solid and liquid biopsies provides additional clinical information to improve patient care. J Cancer Metastasis Treat. 2018, 4, null–null. [Google Scholar] [CrossRef]
  312. Kus, T.; Aktas, G.; Oktay, C.; Puyan, F.O.; Tastekin, E. Dramatic response to crizotinib in a breast cancer patient with ALK gene rearrangement. Anti-Cancer Drugs. 2022, 33, 400–5. [Google Scholar] [CrossRef]
  313. Arbab, A.S.; Rashid, M.H.; Angara, K.; Borin, T.F.; Lin, P.C.; Jain, M.; et al. Major Challenges and Potential Microenvironment-Targeted Therapies in Glioblastoma. Int J Mol Sci. 2017, 18, 2732. [Google Scholar] [CrossRef] [PubMed]
  314. Zhang, Y.; Coleman, M.; Brekken, R.A. Perspectives on Hypoxia Signaling in Tumor Stroma. Cancers. 2021, 13, 3070. [Google Scholar] [CrossRef]
  315. Jr Jp Polivka, J.; Holubec, L.; Kubikova, T.; Priban, V.; Hes, O.; et al. Advances in Experimental Targeted Therapy and Immunotherapy for Patients with Glioblastoma Multiforme. Anticancer Res. 2017, 37, 21–33. [Google Scholar] [CrossRef] [PubMed]
  316. Letai, A.; Bhola, P.; Welm, A.L. Functional precision oncology: Testing tumors with drugs to identify vulnerabilities and novel combinations. Cancer Cell. 2022, 40, 26–35. [Google Scholar] [CrossRef]
  317. Ho HY, Chung KS (Kasey), Kan CM, Wong SC (Cesar). Liquid Biopsy in the Clinical Management of Cancers. Int J Mol Sci. 2024, 25, 8594. [Google Scholar] [CrossRef]
  318. Noor, J.; Chaudhry, A.; Noor, R.; Batool, S. Advancements and Applications of Liquid Biopsies in Oncology: A Narrative Review. Cureus. 2023, 15, e42731. [Google Scholar] [CrossRef] [PubMed]
  319. Gumà, J.; Peña, K.; Riu, F.; Guilarte, C.; Hernandez, A.; Lucía, C.; et al. Utility of ctDNA Liquid Biopsies from Cancer Patients: An Institutional Study of 285 ctDNA Samples. Cancers. 2022, 14, 5859. [Google Scholar] [CrossRef] [PubMed]
  320. Li, D.; Lai, W.; Fan, D.; Fang, Q. Protein biomarkers in breast cancer-derived extracellular vesicles for use in liquid biopsies. Am J Physiol-Cell Physiol. 2021, 321, C779–97. [Google Scholar] [CrossRef]
  321. Han, H.S.; Lee, K.W. Liquid Biopsy: An Emerging Diagnostic, Prognostic, and Predictive Tool in Gastric Cancer. J Gastric Cancer. 2024, 24, 4–28. [Google Scholar] [CrossRef]
  322. Sehayek, O.; Kian, W.; Onn, A.; Stoff, R.; Sorotsky, H.G.; Zemel, M.; et al. Liquid First Is “Solid” in Naïve Non-Small Cell Lung Cancer Patients: Faster Turnaround Time With High Concordance to Solid Next-Generation Sequencing. Front Oncol. 2022, 12, 912801. [Google Scholar] [CrossRef]
  323. Saarenheimo, J.; Eigeliene, N.; Andersen, H.; Tiirola, M.; Jekunen, A. The Value of Liquid Biopsies for Guiding Therapy Decisions in Non-small Cell Lung Cancer. Front Oncol. 2019, 9, 129. [Google Scholar] [CrossRef]
  324. Wang, H.; Zhang, Y.; Zhang, H.; Cao, H.; Mao, J.; Chen, X.; et al. Liquid biopsy for human cancer: cancer screening, monitoring, and treatment. MedComm. 2024, 5, e564. [Google Scholar] [CrossRef]
  325. Chehade, C.H.; Ozay, Z.I.; Agarwal, N. Targeting the FGFR Pathway in Patients with Advanced Solid Tumors. Clin Cancer Res. 2024, 30, 4549–51. [Google Scholar] [CrossRef] [PubMed]
  326. Hu-Lieskovan, S.; Malouf, G.G.; Jacobs, I.; Chou, J.; Liu, L.; Johnson, M.L. Addressing resistance to immune checkpoint inhibitor therapy: an urgent unmet need. Futur Oncol. 2021, 17, 1401–39. [Google Scholar] [CrossRef]
  327. Kasim, A.; Bean, N.; Hendriksen, S.J.; Chen, T.T.; Zhou, H.; Psioda, M.A. Basket trials in oncology: a systematic review of practices and methods, comparative analysis of innovative methods, and an appraisal of a missed opportunity. Front Oncol. 2023, 13, 1266286. [Google Scholar] [CrossRef]
  328. Iyer, G.; Kwiatkowski, D.J.; Ding, L.; Schmid, A.N.; Navarro, W.H.; Ahnert, J.R. PRECISION 1: A phase 2, multicenter, open-label basket trial of nab-sirolimus for malignant solid tumors harboring pathogenic inactivating alterations in TSC1 and TSC2. J Clin Oncol. 2024;42(4_suppl):TPS526–TPS526.
  329. Raj, N.; Zheng, Y.; Kelly, V.; Katz, S.S.; Chou, J.; Do, R.K.G.; et al. PD-1 Blockade in Advanced Adrenocortical Carcinoma. J Clin Oncol. 2020, 38, 71–80. [Google Scholar] [CrossRef]
  330. Deleuze, A.; Saout, J.; Dugay, F.; Peyronnet, B.; Mathieu, R.; Verhoest, G.; et al. Immunotherapy in Renal Cell Carcinoma: The Future Is Now. Int J Mol Sci. 2020, 21, 2532. [Google Scholar] [CrossRef] [PubMed]
  331. Juang, H.H.; Chen, S.M.; Lin, G.; Chiang, M.H.; Hou, C.P.; Lin, Y.H.; et al. The Clinical Experiences of Urine Metabolomics of Genitourinary Urothelial Cancer in a Tertiary Hospital in Taiwan. Front Oncol. 2021, 11, 680910. [Google Scholar] [CrossRef] [PubMed]
  332. Liu, X.; He, M.; Li, L.; Wang, X.; Han, S.; Zhao, J.; et al. EMT and Cancer Cell Stemness Associated With Chemotherapeutic Resistance in Esophageal Cancer. Front Oncol. 2021, 11, 672222. [Google Scholar] [CrossRef]
  333. Adam-Zahir, S.; Plowman, P.N.; Bourton, E.C.; Sharif, F.; Parris, C.N. Increased γ-H2AX and Rad51 DNA Repair Biomarker Expression in Human Cell Lines Resistant to the Chemotherapeutic Agents Nitrogen Mustard and Cisplatin. Chemotherapy.
  334. Ma, S.; Zhou, M.; Xu, Y.; Gu, X.; Zou, M.; Abudushalamu, G.; et al. Clinical application and detection techniques of liquid biopsy in gastric cancer. Mol Cancer. 2023, 22, 7. [Google Scholar] [CrossRef] [PubMed]
  335. Johann, D.J.; Steliga, M.; Shin, I.J.; Yoon, D.; Arnaoutakis, K.; Hutchins, L.; et al. Liquid biopsy and its role in an advanced clinical trial for lung cancer. Exp Biol Med. 2018, 243, 262–71. [Google Scholar] [CrossRef]
  336. Carlo Ed Schiappacassi, M.; Pelizzari, G.; Baresic, T.; Conte Ad Stanzione, B.; et al. Acquired EGFR C797G Mutation Detected by Liquid Biopsy as Resistance Mechanism After Treatment With Osimertinib: A Case Report. Vivo. 2021, 35, 2941–5. [Google Scholar] [CrossRef]
  337. Durgut, S.; Salihefendić, L.; Pećar, D.; Čeko, I.; Mulahuseinović, N.; Izmirlija, M.; et al. Droplet Digital PCR as a Molecular Tool for the Detection of the EGFR T790M Mutation in NSCLC Patients with the EGFR Activating Mutations. Balk J Méd Genet. 2023, 26, 21–6. [Google Scholar] [CrossRef]
  338. Lu, J.; Han, B. Liquid Biopsy Promotes Non-Small Cell Lung Cancer Precision Therapy. Technol Cancer Res Treat. 2018, 17, 1533033818801809. [Google Scholar] [CrossRef]
  339. Lau, C.; Jamali, F.; Loebenberg, R. Health Canada Usage of Real World Evidence (RWE) in Regulatory Decision Making compared with FDA/EMA usage based on publicly available information: Real-World Evidence used by Health Canada in Regulatory Decision Making. J Pharm Pharm Sci. 2022, 25, 227–36. [Google Scholar] [CrossRef]
  340. Jackson, M.L.; Manickam, R.; Derieg, D.; Gombar, S.; Low, Y.S. Quantifying Fit-for-Purpose in Real World Data: Data Grading and FitQ Scores. medRxiv. 2430. [Google Scholar]
  341. Khozin, S.; Abernethy, A.P.; Nussbaum, N.C.; Zhi, J.; Curtis, M.D.; Tucker, M.; et al. Characteristics of Real-World Metastatic Non-Small Cell Lung Cancer Patients Treated with Nivolumab and Pembrolizumab During the Year Following Approval. Oncol. 2018, 23, 328–36. [Google Scholar] [CrossRef]
  342. Green, A.K.; Curry, M.; Trivedi, N.; Bach, P.B.; Mailankody, S. Assessment of Outcomes Associated With the Use of Newly Approved Oncology Drugs in Medicare Beneficiaries. JAMA Netw Open. 2021, 4, e210030. [Google Scholar] [CrossRef] [PubMed]
  343. Mahal, B.A.; Chen, Y.; Muralidhar, V.; Mahal, A.R.; Choueiri, T.K.; Hoffman, K.E.; et al. National sociodemographic disparities in the treatment of high-risk prostate cancer: Do academic cancer centers perform better than community cancer centers? Cancer. 2016, 122, 3371–7. [Google Scholar] [CrossRef]
  344. Barocas, D.A.; Gray, D.T.; Fowke, J.H.; Mercaldo, N.D.; Blume, J.D.; Chang, S.S.; et al. Racial Variation in the Quality of Surgical Care for Prostate Cancer. J Urol. 2012, 188, 1279–85. [Google Scholar] [CrossRef] [PubMed]
  345. Torre, L.A.; Sauer, A.M.G.; Chen, M.S.; Kagawa-Singer, M.; Jemal, A.; Siegel, R.L. Cancer statistics for Asian Americans, Native Hawaiians, and Pacific Islanders, 2016: Converging incidence in males and females. CA: A Cancer J Clin. 2016, 66, 182–202. [Google Scholar] [CrossRef]
  346. Ryoo, J.J.; Ordin, D.L.; Antonio, A.L.M.; Oishi, S.M.; Gould, M.K.; Asch, S.M.; et al. Patient Preference and Contraindications in Measuring Quality of Care: What Do Administrative Data Miss? J Clin Oncol. 2013, 31, 2716–23. [Google Scholar] [CrossRef]
  347. Wolfson, J.A.; Sun, C.L.; Kim, H.; Kang, T.; Bhatia, S. Evaluation of the effect of care at NCI comprehensive cancer centers (NCICCCs) on disparities in outcome within adolescents and young adults (AYAs) with cancer. J Clin Oncol. 9512. [Google Scholar]
  348. Vingiani, A.; Agnelli, L.; Duca, M.; Lorenzini, D.; Damian, S.; Proto, C.; et al. Molecular Tumor Board as a Clinical Tool for Converting Molecular Data Into Real-World Patient Care. JCO Precis Oncol. 2023, 7, e2300067. [Google Scholar] [CrossRef] [PubMed]
  349. Schwaederle, M.; Parker, B.A.; Schwab, R.B.; Fanta, P.T.; Boles, S.G.; Daniels, G.A.; et al. Molecular Tumor Board: The University of California San Diego Moores Cancer Center Experience. Oncol. 2014, 19, 631–6. [Google Scholar] [CrossRef]
  350. Specchia, M.L.; Frisicale, E.M.; Carini, E.; Pilla, A.D.; Cappa, D.; Barbara, A.; et al. The impact of tumor board on cancer care: evidence from an umbrella review. BMC Heal Serv Res. 2020, 20, 73. [Google Scholar] [CrossRef]
  351. Pujol, P.; Rouge, T.D.L.M.; Penault-Llorca, F. From Targeting Somatic Mutations to Finding Inherited Cancer Predispositions: The Other Side of the Coin. Diagnostics. 2019, 9, 83. [Google Scholar] [CrossRef]
  352. Salgia, R. Diagnostic challenges in non-small-cell lung cancer: an integrated medicine approach. Futur Oncol. 2015, 11, 489–500. [Google Scholar] [CrossRef] [PubMed]
  353. Tan, D.S.W.; Yom, S.S.; Tsao, M.S.; Pass, H.I.; Kelly, K.; Peled, N.; et al. The International Association for the Study of Lung Cancer Consensus Statement on Optimizing Management of EGFR Mutation–Positive Non–Small Cell Lung Cancer: Status in 2016. J Thorac Oncol. 2016, 11, 946–63. [Google Scholar] [CrossRef]
  354. Broes, S.; Saesen, R.; Lacombe, D.; Huys, I. Past, Current, and Future Cancer Clinical Research Collaborations: The Case of the European Organisation for Research and Treatment of Cancer. Clin Transl Sci. 2020, 14, 47–53. [Google Scholar] [CrossRef] [PubMed]
  355. Patel, A.C.; Coyle, A.J. Building a New Biomedical Ecosystem: Pfizer’s Centers for Therapeutic Innovation. Clin Pharmacol Ther. 2013, 94, 314–6. [Google Scholar] [CrossRef]
  356. Stahel, R.A.; Lacombe, D.; Cardoso, F.; Casali, P.G.; Negrouk, A.; Marais, R.; et al. Current models, challenges and best practices for work conducted between European academic cooperative groups and industry. ESMO Open. 2020, 5, e000628. [Google Scholar] [CrossRef] [PubMed]
  357. Kasichayanula, S.; Mandlekar, S.; Shivva, V.; Patel, M.; Girish, S. Evolution of preclinical characterization and insights into clinical pharmacology of checkpoint inhibitors approved for cancer immunotherapy. Clin Transl Sci. 2022, 15, 1818–37. [Google Scholar] [CrossRef]
  358. Palafox, N.; Guerrero, R.L.; Robinett, H.; Peterson, J.; Ward, D.; Vogel, C.W. Advancing Cancer Health Equity in Pacific Islanders: A 15-Year Investment in Cancer Research, Training and Outreach in Guam, Hawaii and the U.S. Associated Pacific Islands. J Glob Oncol. 1: 2).
  359. Guerrero, R.T.L.; Hattori-Uchima, M.; Robinett, H.R.; Vogel, C.W.; Palafox, N.A. Abstract PO-036: University of Guam/University of Hawai‘i Cancer Center Partnership: A seventeen-year investment in cancer health equity in Pacific Islanders. Cancer Epidemiology, Biomark Prev. 2020;29(12_Supplement):PO-036-PO-036.
  360. Mazariego, C.; Daly, R.; McGill, B.; Kelada, L.; McKay, S.; Hetherington, K.; et al. Barriers to access of precision guided therapies for children with high-risk cancer. Pediatr Blood Cancer. 2024, 71, e31147. [Google Scholar] [CrossRef]
  361. Moerdler, S.; Zhang, L.; Gerasimov, E.; Zhu, C.; Wolinsky, T.; Roth, M.; et al. Physician perspectives on compassionate use in pediatric oncology. Pediatr Blood Cancer. 2018, 66, e27545. [Google Scholar] [CrossRef]
  362. Tan, A.C.; Bagley, S.J.; Wen, P.Y.; Lim, M.; Platten, M.; Colman, H.; et al. Systematic review of combinations of targeted or immunotherapy in advanced solid tumors. J Immunother Cancer. 2021, 9, e002459. [Google Scholar] [CrossRef]
  363. Moffit, J.S.; Blanset, D.L.; Lynch, J.L.; MacLachlan, T.K.; Meyer, K.E.; Ponce, R.; et al. Regulatory Consideration for the Nonclinical Safety Assessment of Gene Therapies. Hum Gene Ther. 1126. [Google Scholar]
  364. Stride, E.; Segers, T.; Lajoinie, G.; Cherkaoui, S.; Bettinger, T.; Versluis, M.; et al. Microbubble Agents: New Directions. Ultrasound Med Biol. 2020, 46, 1326–43. [Google Scholar] [CrossRef]
  365. Mifsud, J.; Cranswick, N. Addressing the challenges of novel therapies in rare diseases with mechanistic perspectives: Missed opportunities or the way forward? Br J Clin Pharmacol. 2022, 88, 2480–3. [Google Scholar] [CrossRef] [PubMed]
  366. Castellano, D.; Apolo, A.B.; Porta, C.; Capdevila, J.; Viteri, S.; Rodriguez-Antona, C.; et al. Cabozantinib combination therapy for the treatment of solid tumors: a systematic review. Ther Adv Méd Oncol. 2022, 14, 17588359221108692. [Google Scholar] [CrossRef]
  367. Bansal, D.; Reimers, M.A.; Knoche, E.M.; Pachynski, R.K. Immunotherapy and Immunotherapy Combinations in Metastatic Castration-Resistant Prostate Cancer. Cancers. 2021, 13, 334. [Google Scholar] [CrossRef]
  368. Benjamin, D.J.; Padula, W.V.; Hsu, R.C. Cost effectiveness of immunotherapy combination therapies for endometrial cancer. Gynecol Oncol Rep. 2024, 52, 101351. [Google Scholar] [CrossRef]
  369. Dranitsaris, G.; Zhu, X.; Adunlin, G.; Vincent, M.D. Cost effectiveness vs. affordability in the age of immuno-oncology cancer drugs. Expert Rev Pharmacoeconomics Outcomes Res. 2018, 18, 351–7. [Google Scholar] [CrossRef] [PubMed]
  370. Huang, L.Y.; Gau, C.S. Lessons learned from the reimbursement policy for immune checkpoint inhibitors and real-world data collection in Taiwan. Int J Technol Assess Heal Care. 2020, 37, e26. [Google Scholar] [CrossRef]
  371. Fokas, E.; Appelt, A.; Glynne-Jones, R.; Beets, G.; Perez, R.; Garcia-Aguilar, J.; et al. International consensus recommendations on key outcome measures for organ preservation after (chemo)radiotherapy in patients with rectal cancer. Nat Rev Clin Oncol. 2021, 18, 805–16. [Google Scholar] [CrossRef]
  372. Sung, P.S. Crosstalk between tumor-associated macrophages and neighboring cells in hepatocellular carcinoma. Clin Mol Hepatol. 2022, 28, 333–50. [Google Scholar] [CrossRef]
  373. Twomey, J.D.; Zhang, B. Cancer Immunotherapy Update: FDA-Approved Checkpoint Inhibitors and Companion Diagnostics. AAPS J. 2021, 23, 39. [Google Scholar] [CrossRef]
  374. Roberts, N.A.; Dhillon, H.M.; Paterson, C.; Schubach, K.; McJannett, M.; Group, A. ; NZU; PCT The impact of coronavirus disease 2019 on genitourinary and prostate cancer care and clinical trials: A qualitative exploration of the Australian and New Zealand experience. AsiaPac J Clin Oncol. 2022, 19, 337–46. [Google Scholar]
  375. Li, L.; Sivasankaran, G.; Wijayawardena, B.K. Heterogeneity in untreated, stressed and drug-tolerant cells: insights into the evolution of cancer resistance. Int J Comput Biol Drug Des.
  376. Wang, Q.; Guldner, I.H.; Golomb, S.M.; Sun, L.; Harris, J.; Lu, X.; et al. Single-cell profiling guided combinatorial immunotherapy for fast-evolving CDK4/6 inhibitor resistant HER2-positive breast cancer. bioRxiv. 1198. [Google Scholar]
  377. Emert, B.L.; Cote, C.; Torre, E.A.; Dardani, I.P.; Jiang, C.L.; Jain, N.; et al. Variability within rare cell states enables multiple paths towards drug resistance. bioRxiv. 9966. [Google Scholar]
  378. Boolchandani, M.; D’Souza, A.W.; Dantas, G. Sequencing-based methods and resources to study antimicrobial resistance. Nat Rev Genet. 2019, 20, 356–70. [Google Scholar] [CrossRef] [PubMed]
  379. Xu, S.; Liu, M.; Bai, Y.; Liu, H. Multi-Dimensional Organic Mass Cytometry: Simultaneous Analysis of Proteins and Metabolites on Single Cells. Angew Chem Int Ed. 2020, 60, 1806–12. [Google Scholar] [CrossRef]
  380. Fan, X.X.; Wu, Q. Decoding Lung Cancer at Single-Cell Level. Front Immunol. 2022, 13, 883758. [Google Scholar] [CrossRef]
  381. Lawson, D.A.; Kessenbrock, K.; Davis, R.T.; Pervolarakis, N.; Werb, Z. Tumour heterogeneity and metastasis at single-cell resolution. Nat Cell Biol. 2018, 20, 1349–60. [Google Scholar] [CrossRef]
  382. Schnepp, P.M.; Shelley, G.; Dai, J.; Wakim, N.; Jiang, H.; Mizokami, A.; et al. Single-Cell Transcriptomics Analysis Identifies Nuclear Protein 1 as a Regulator of Docetaxel Resistance in Prostate Cancer Cells. Mol Cancer Res. 2020, 18, 1290–301. [Google Scholar] [CrossRef]
  383. Lin, P.; Cheng, W.; Qi, X.; Zhang, P.; Xiong, J.; Li, J. Bioinformatics and Experimental Validation for Identifying Biomarkers Associated with AMG510 (Sotorasib) Resistance in KRASG12C-Mutated Lung Adenocarcinoma. Int J Mol Sci. 2024, 25, 1555. [Google Scholar] [CrossRef]
  384. Chiribau, C.B.; Schmedes, S.; Dong, Y.; Tarigopula, N.; Tekin, O.; Cannons, A.; et al. Detection of resistance to macrolides and fluoroquinolones in Mycoplasma genitalium by targeted next-generation sequencing. Microbiol Spectr. 2024, 12, e03845–23. [Google Scholar] [CrossRef] [PubMed]
  385. Guo, J.; Sun, D.; Li, K.; Dai, Q.; Geng, S.; Yang, Y.; et al. Metabolic Labeling and Digital Microfluidic Single-Cell Sequencing for Single Bacterial Genotypic-Phenotypic Analysis. Small. 2024, 20, e2402177. [Google Scholar] [CrossRef] [PubMed]
  386. Gomes, B.C.; Honrado, M.; Armada, A.; Viveiros, M.; Rueff, J.; Rodrigues, A.S. ABC Efflux Transporters and the Circuitry of miRNAs: Kinetics of Expression in Cancer Drug Resistance. Int J Mol Sci. 2020, 21, 2985. [Google Scholar] [CrossRef]
  387. Zhu, G.; Zhang, W.; Zhao, Y.; Wang, G.; Yuan, H.; Guo, G.; et al. Single-Cell Mass Spectrometry Studies of Secondary Drug Resistance of Tumor Cells. Anal Chem. 2024, 97, 337–44. [Google Scholar] [CrossRef]
  388. Sun, M.; Chen, X.; Yang, Z. Single cell mass spectrometry studies reveal metabolomic features and potential mechanisms of drug-resistant cancer cell lines. Anal Chim Acta. 2022, 1206, 339761. [Google Scholar] [CrossRef]
  389. Champagne, A.; Jain, P.; Vélot, L.; Riopel, J.; Lefebvre, V.; Neveu, B.; et al. A transcriptional biosensor to monitor single cancer cell therapeutic responses by bioluminescence microscopy. Theranostics. 2022, 12, 474–92. [Google Scholar] [CrossRef] [PubMed]
  390. Toth, R.; Schiffmann, H.; Hube-Magg, C.; Büscheck, F.; Höflmayer, D.; Weidemann, S.; et al. Random forest-based modelling to detect biomarkers for prostate cancer progression. Clin Epigenetics. 2019, 11, 148. [Google Scholar] [CrossRef]
  391. Alghafees, M.; Seyam, R.M.; Al-Hussain, T.; Amin, T.M.; Altaweel, W.; Sabbah, B.N.; et al. Using machine learning models to predict synchronous genitourinary cancers among gastrointestinal stromal tumor patients. Urol Ann. 2024, 16, 94–7. [Google Scholar] [CrossRef] [PubMed]
  392. Khagi, Y.; Goodman, A.M.; Daniels, G.A.; Patel, S.P.; Sacco, A.G.; Randall, J.M.; et al. Hypermutated Circulating Tumor DNA: Correlation with Response to Checkpoint Inhibitor–Based Immunotherapy. Clin Cancer Res. 2017, 23, 5729–36. [Google Scholar] [CrossRef] [PubMed]
  393. Ivanova, E.; Asadullina, D.; Gilyazova, G.; Rakhimov, R.; Izmailov, A.; Pavlov, V.; et al. Exosomal MicroRNA Levels Associated with Immune Checkpoint Inhibitor Therapy in Clear Cell Renal Cell Carcinoma. Biomedicines. 2023, 11, 801. [Google Scholar] [CrossRef]
  394. Kelloff, G.J.; Sigman, C.C.; Scher, H.I. Biomarker development in the context of urologic cancers. Urol Oncol: Semin Orig Investig. 2015, 33, 295–301. [Google Scholar] [CrossRef]
  395. Hugen, C.M.; Zainfeld, D.E.; Goldkorn, A. Circulating Tumor Cells in Genitourinary Malignancies: An Evolving Path to Precision Medicine. Front Oncol. 2017, 7, 6. [Google Scholar] [CrossRef]
  396. Polley, M.Y.C.; Cheung, Y.K. Early-Phase Platform Trials: A New Paradigm for Dose Finding and Treatment Screening in the Era of Precision Oncology. JCO Precis Oncol. 2019, 3, 1–8. [Google Scholar] [CrossRef]
  397. Hu, C.; Dignam, J.J. Biomarker-Driven Oncology Clinical Trials: Key Design Elements, Types, Features, and Practical Considerations. JCO Precis Oncol. 2019, 3, 1–12. [Google Scholar] [CrossRef]
  398. Zhou, Q.; Chen, H.J.; Wang, B.C.; Wang, Z.; Tu, H.Y.; Xu, C.; et al. CLUSTER: A biomarker-integrated targeted therapy study in patients with advanced non-small cell lung cancer. 2022. [Google Scholar]
  399. Lee Ckun Kim, H.S.; Jung, M.; Kim, H.; Bae, W.K.; Koo, D.H.; et al. Open-Label, Multicenter, Randomized, Biomarker-Integrated Umbrella Trial for Second-Line Treatment of Advanced Gastric Cancer: K-Umbrella Gastric Cancer Study. J Clin Oncol. 2024, 42, 348–57. [Google Scholar]
  400. Wu, S.; Thawani, R. Tumor-Agnostic Therapies in Practice: Challenges, Innovations, and Future Perspectives. Cancers. 2025, 17, 801. [Google Scholar] [CrossRef]
  401. Burd, A.; Schilsky, R.L.; Byrd, J.C.; Levine, R.L.; Papadimitrakopoulou, V.A.; Herbst, R.S.; et al. Challenges and approaches to implementing master/basket trials in oncology. Blood Adv. 2019, 3, 2237–43. [Google Scholar] [CrossRef]
  402. van de Wetering, M.; Francies, H.E.; Francis, J.M.; Bounova, G.; Iorio, F.; Pronk, A.; et al. Prospective Derivation of a Living Organoid Biobank of Colorectal Cancer Patients . Cell 2015, 161, 933–45. [Google Scholar] [CrossRef]
  403. Pappas, K.J.; Choi, D.; Sawyers, C.L.; Karthaus, W.R. Prostate Organoid Cultures as Tools to Translate Genotypes and Mutational Profiles to Pharmacological Responses. J Vis Exp.
  404. Kijima, T.; Nakagawa, H.; Shimonosono, M.; Chandramouleeswaran, P.M.; Hara, T.; Sahu, V.; et al. Three-Dimensional Organoids Reveal Therapy Resistance of Esophageal and Oropharyngeal Squamous Cell Carcinoma Cells. Cell Mol Gastroenterol Hepatol. 2019, 7, 73–91. [Google Scholar] [CrossRef] [PubMed]
  405. Lee, S.H.; Hu, W.; Matulay, J.T.; Silva, M.V.; Owczarek, T.B.; Kim, K.; et al. Tumor Evolution and Drug Response in Patient-Derived Organoid Models of Bladder Cancer. Cell. 2018, 173, 515–528. [Google Scholar] [CrossRef]
  406. Beshiri, M.L.; Tice, C.M.; Tran, C.; Nguyen, H.M.; Sowalsky, A.G.; Agarwal, S.; et al. A PDX/organoid biobank of advanced prostate cancers captures genomic and phenotypic heterogeneity for disease modeling and therapeutic screening. Clin Cancer Res. 2018, 24, clincanres. [Google Scholar] [CrossRef]
  407. Cao, Q.; Li, L.; Zhao, Y.; Wang, C.; Shi, Y.; Tao, X.; et al. PARPi Decreased Primary Ovarian Cancer Organoid Growth Through Early Apoptosis and Base Excision Repair Pathway. Cell Transplant. 2023, 32, 09636897231187996. [Google Scholar] [CrossRef] [PubMed]
  408. Vlachogiannis, G.; Hedayat, S.; Vatsiou, A.; Jamin, Y.; Fernández-Mateos, J.; Khan, K.; et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science. 2018, 359, 920–6. [Google Scholar] [CrossRef]
  409. Ziani, L.; Buart, S.; Chouaib, S.; Thiery, J. Hypoxia increases melanoma-associated fibroblasts immunosuppressive potential and inhibitory effect on T cell-mediated cytotoxicity. OncoImmunology. 2021, 10, 1950953. [Google Scholar] [CrossRef]
  410. Wang, Z.; Liang, X.; Zhang, H.; Wang, Z.; Zhang, X.; Dai, Z.; et al. Identification of a Hypoxia-Angiogenesis lncRNA Signature Participating in Immunosuppression in Gastric Cancer. J Immunol Res. 2022, 2022, 5209607. [Google Scholar] [CrossRef] [PubMed]
  411. Gnanamony, M.; Demirkhanyan, L.; Ge, L.; Sojitra, P.; Bapana, S.; Norton, J.A.; et al. Circular dumbbell miR-34a-3p and −5p suppresses pancreatic tumor cell-induced angiogenesis and activates macrophages. Oncol Lett. 2020, 21, 75. [Google Scholar] [CrossRef] [PubMed]
  412. Li, M.; He, L.; Zhu, J.; Zhang, P.; Liang, S. Targeting tumor-associated macrophages for cancer treatment. Cell Biosci. 2022, 12, 85. [Google Scholar] [CrossRef]
  413. Takahashi, H.; Rokudai, S.; Kawabata-Iwakawa, R.; Sakakura, K.; Oyama, T.; Nishiyama, M.; et al. AKT3 Is a Novel Regulator of Cancer-Associated Fibroblasts in Head and Neck Squamous Cell Carcinoma. Cancers. 2021, 13, 1233. [Google Scholar] [CrossRef]
  414. Fukumura, D.; Kloepper, J.; Amoozgar, Z.; Duda, D.G.; Jain, R.K. Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nat Rev Clin Oncol. 2018, 15, 325–40. [Google Scholar] [CrossRef]
  415. Lopes-Coelho, F.; Martins, F.; Pereira, S.A.; Serpa, J. Anti-Angiogenic Therapy: Current Challenges and Future Perspectives. Int J Mol Sci. 2021, 22, 3765. [Google Scholar] [CrossRef]
  416. Song, Y.; Fu, Y.; Xie, Q.; Zhu, B.; Wang, J.; Zhang, B. Anti-angiogenic Agents in Combination With Immune Checkpoint Inhibitors: A Promising Strategy for Cancer Treatment. Front Immunol. 2020, 11, 1956. [Google Scholar] [CrossRef] [PubMed]
  417. Lee, W.S.; Yang, H.; Chon, H.J.; Kim, C. Combination of anti-angiogenic therapy and immune checkpoint blockade normalizes vascular-immune crosstalk to potentiate cancer immunity. Exp Mol Med. 2020, 52, 1475–85. [Google Scholar] [CrossRef]
  418. Chang, C.C.; Dinh, T.K.; Lee, Y.A.; Wang, F.N.; Sung, Y.C.; Yu, P.L.; et al. Nanoparticle Delivery of MnO2 and Antiangiogenic Therapy to Overcome Hypoxia-Driven Tumor Escape and Suppress Hepatocellular Carcinoma. ACS Appl Mater Interfaces. 2020, 12, 44407–19. [Google Scholar] [CrossRef]
  419. Shamshiripour, P.; Hajiahmadi, F.; Lotfi, S.; Esmaeili, N.R.; Zare, A.; Akbarpour, M.; et al. Next-Generation Anti-Angiogenic Therapies as a Future Prospect for Glioma Immunotherapy; From Bench to Bedside. Front Immunol. 2022, 13, 859633. [Google Scholar] [CrossRef]
  420. Hang, R.; Tian, X.; Qu, G.; Zhao, Y.; Yao, R.; Zhang, Y.; et al. Exosomes derived from magnesium ion—stimulated macrophages inhibit angiogenesis. Biomed Mater. 2022, 17, 045008. [Google Scholar] [CrossRef] [PubMed]
  421. Yang, Q.; Xu, J.; Gu, J.; Shi, H.; Zhang, J.; Zhang, J.; et al. Extracellular Vesicles in Cancer Drug Resistance: Roles, Mechanisms, and Implications. Adv Sci. 2022, 9, 2201609. [Google Scholar] [CrossRef] [PubMed]
  422. Mir, R.; Baba, S.K.; Elfaki, I.; Algehainy, N.; Alanazi, M.A.; Altemani, F.H.; et al. Unlocking the Secrets of Extracellular Vesicles: Orchestrating Tumor Microenvironment Dynamics in Metastasis, Drug Resistance, and Immune Evasion. J Cancer. 2024, 15, 6383–415. [Google Scholar] [CrossRef] [PubMed]
  423. Li, I.; Nabet, B.Y. Exosomes in the tumor microenvironment as mediators of cancer therapy resistance. Mol Cancer. 2019, 18, 32. [Google Scholar] [CrossRef]
  424. Trentham-Dietz, A.; Bird, J.E.; Gangnon, R.E.; Lindberg, S.M.; Madison, T.; Malecki, K.M.C.; et al. Coordinating Centers as a Strategy for Accelerating Cancer Epidemiology Consortia: Best Practices. Curr Epidemiology Rep. 2022, 9, 1–9. [Google Scholar] [CrossRef]
  425. Harris, J.K.; Provan, K.G.; Johnson, K.J.; Leischow, S.J. Drawbacks and benefits associated with inter-organizational collaboration along the discovery-development-delivery continuum: a cancer research network case study. Implement Sci. 2012, 7, 69. [Google Scholar] [CrossRef]
  426. Varmus, H.; Kumar, H.S. Addressing the Growing International Challenge of Cancer: A Multinational Perspective. Sci Transl Med. 2013, 5, 175cm2. [Google Scholar] [CrossRef]
  427. Rowland, J.H.; Kent, E.E.; Forsythe, L.P.; Loge, J.H.; Hjorth, L.; Glaser, A.; et al. Cancer survivorship research in Europe and the United States: Where have we been, where are we going, and what can we learn from each other? Cancer. 2013;119(S11):2094–108.
  428. Froggatt, K.; Preston, N.; Turner, M.; Kerr, C. Patient and public involvement in research and the Cancer Experiences Collaborative: benefits and challenges. BMJ Support Palliat Care. 2014, 5, 518. [Google Scholar] [CrossRef]
  429. Vogel, A.L.; Perin, D.M.P.; Lu, Y.L.; Taplin, S.H. Understanding the Value of International Research Networks: An Evaluation of the International Cancer Screening Network of the US National Cancer Institute. J Glob Oncol. 2019, 5, JGO. [Google Scholar] [CrossRef]
  430. Pal, T.; Suiter, S.V.; Moses, H.L.; Smoot, D.T.; Richmond, A.; Tiriveedhi, V.; et al. The Meharry-Vanderbilt-Tennessee State University Cancer Partnership (MVTCP): History and Highlights of 20 Years of Accomplishments. J Heal Care Poor Underserved. 2022, 33, 419–36. [Google Scholar] [CrossRef]
  431. Banydeen, R.; Rose, A.M.C.; Martin, D.; Aiken, W.; Alexis, C.; Andall-Brereton, G.; et al. Advancing Cancer Control through Research and Cancer Registry Collaborations in the Caribbean. Cancer Control. 2015, 22, 520–30. [Google Scholar] [CrossRef] [PubMed]
  432. Celis, J.E.; Heitor, M. Towards a mission-oriented approach to cancer in Europe: an unmet need in cancer research policy. Mol Oncol. 2019, 13, 502–10. [Google Scholar] [CrossRef]
  433. Berns, A. Quality-assured research environments for translational cancer research. Mol Oncol. 2019, 13, 543–8. [Google Scholar] [CrossRef]
  434. Brokowski, C.; Adli, M. CRISPR Ethics: Moral Considerations for Applications of a Powerful Tool. J Mol Biol. 2019, 431, 88–101. [Google Scholar] [CrossRef] [PubMed]
  435. Chaudhry, A.T.; Akhtar, D. Gene Therapy and Modification as a Therapeutic Strategy for Cancer. Univ Ott J Med. 2016, 6, 44–8. [Google Scholar] [CrossRef]
  436. Kolanu, N.D. CRISPR–Cas9 Gene Editing: Curing Genetic Diseases by Inherited Epigenetic Modifications. Glob Méd Genet. 2024, 11, 113–22. [Google Scholar] [CrossRef]
  437. Ikwuka, A.O.; Musa, S.; Udeh, F.C.; Musa, A.A.; Chukwuezie, U.C. CRISPR-Cas9 Genomic Editing as an Innovation in the Management of Sickle Cell Disease: A Systematic Review. Am J Méd Sci Innov. 2023, 2, 36–48. [Google Scholar] [CrossRef]
  438. Schweikart, S.J. What Is Prudent Governance of Human Genome Editing? AMA J Ethics. 2019, 21, E1042–1048. [Google Scholar]
  439. SRK; Kotian, H.; Chatterjee, P.K.; Yuguda, Y.M.; Rochmat, A.; To, T.T.H. SRK; Kotian, H.; Chatterjee, P.K.; Yuguda, Y.M.; Rochmat, A.; To, T.T.H.; et al. Ethical Implications And Molecular Mechanisms Of CRISPR-Cas9 In Modern Biology. Afr J Biomed Res. 1520. [Google Scholar]
  440. Guo, N.; Liu, J.B.; Li, W.; Ma, Y.S.; Fu, D. The power and the promise of CRISPR/Cas9 genome editing for clinical application with gene therapy. J Adv Res. 2022, 40, 135–52. [Google Scholar] [CrossRef]
  441. Dzilic, E.; Lahm, H.; Dreßen, M.; Deutsch, M.A.; Lange, R.; Wu, S.M.; et al. Genome Editing Redefines Precision Medicine in the Cardiovascular Field. Stem Cells Int. 2018, 2018, 4136473. [Google Scholar] [CrossRef]
  442. Ledzewicz, U.; Schättler, H.; Wang, S. On the role of tumor heterogeneity for optimal cancer chemotherapy. Netw Heterog Media. 2019, 14, 131–47. [Google Scholar] [CrossRef]
  443. Karai, E.; Szebényi, K.; Windt, T.; Fehér, S.; Szendi, E.; Dékay, V.; et al. Celecoxib Prevents Doxorubicin-Induced Multidrug Resistance in Canine and Mouse Lymphoma Cell Lines. Cancers. 2020, 12, 1117. [Google Scholar] [CrossRef]
  444. Thomas, D.S.; Cisneros, L.H.; Anderson, A.R.A.; Maley, C.C. In Silico Investigations of Multi-Drug Adaptive Therapy Protocols. Cancers. 2022, 14, 2699. [Google Scholar] [CrossRef]
  445. Piretto, E.; Delitala, M.; Ferraro, M. How Combination Therapies Shape Drug Resistance in Heterogeneous Tumoral Populations. Lett Biomath.
  446. Pan, Y.; Shu, G.; Fu, L.; Huang, K.; Zhou, X.; Gui, C.; et al. EHBP1L1 Drives Immune Evasion in Renal Cell Carcinoma through Binding and Stabilizing JAK1. Adv Sci. 2023, 10, 2206792. [Google Scholar] [CrossRef]
  447. Baghy, K.; Ladányi, A.; Reszegi, A.; Kovalszky, I. Insights into the Tumor Microenvironment—Components, Functions and Therapeutics. Int J Mol Sci. 2023, 24, 17536. [Google Scholar] [CrossRef] [PubMed]
  448. Tang, B.; Wu, C.; Mao, Y.; Qi, X.; Wang, X.; Li, P.; et al. Tracking interactions between TAMs and CAFs mediated by arginase-induced proline production during immune evasion of HCC. 2024. [Google Scholar]
  449. Tanaka, M.; Siemann, D.W. Gas6/Axl Signaling Pathway in the Tumor Immune Microenvironment. Cancers. 2020, 12, 1850. [Google Scholar] [CrossRef] [PubMed]
  450. Nguyen, D.J.M.; Theodoropoulos, G.; Li, Y.Y.; Wu, C.; Sha, W.; Feun, L.G.; et al. Targeting the Kynurenine Pathway for the Treatment of Cisplatin-Resistant Lung Cancer. Mol Cancer Res. 2020, 18, 105–17. [Google Scholar] [CrossRef]
  451. Xu, Y.; He, L.; Fu, Q.; Hu, J. Metabolic Reprogramming in the Tumor Microenvironment With Immunocytes and Immune Checkpoints. Front Oncol. 2021, 11, 759015. [Google Scholar] [CrossRef]
  452. Jiang, M.; Wang, Y.; Zhao, X.; Yu, J. From metabolic byproduct to immune modulator: the role of lactate in tumor immune escape. Front Immunol. 2024, 15, 1492050. [Google Scholar] [CrossRef]
  453. Khan, T.; Nagarajan, M.; Kang, I.; Wu, C.; Wangpaichitr, M. Targeting Metabolic Vulnerabilities to Combat Drug Resistance in Cancer Therapy. J Pers Med. 2025, 15, 50. [Google Scholar] [CrossRef]
  454. George, J.T.; Levine, H. Optimal Cancer Evasion in a Dynamic Immune Microenvironment. bioRxiv. 5027. [Google Scholar]
  455. Dhanasekaran, R.; Hansen, A.S.; Park, J.; Lai, I.; Adeniji, N.; Kuruvilla, S.; et al. MYC Overexpression Drives Immune Evasion in Human Cancer that is Reversible Through Restoration of Pro-Inflammatory Macrophages. bioRxiv. 4918. [Google Scholar]
  456. Jenkins, L.; Jungwirth, U.; Avgustinova, A.; Iravani, M.; Mills, A.P.; Haider, S.; et al. Cancer-associated fibroblasts suppress CD8+ T cell infiltration and confer resistance to immune checkpoint blockade. Cancer Res. 2022, 82, 2904–17. [Google Scholar] [CrossRef] [PubMed]
  457. Liu, X.; Ma, L.; Li, J.; Sun, L.; Yang, Y.; Liu, T.; et al. Trop2-targeted therapies in solid tumors: advances and future directions. Theranostics. 2024, 14, 3674–92. [Google Scholar] [CrossRef]
  458. Bian, X.; Liu, W.; Yang, K.; Sun, C. Therapeutic targeting of PARP with immunotherapy in acute myeloid leukemia. Front Pharmacol. 2024, 15, 1421816. [Google Scholar] [CrossRef] [PubMed]
  459. Sun, C.; Yin, J.; Fang, Y.; Chen, J.; Jeong, K.J.; Chen, X.; et al. BRD4 Inhibition Is Synthetic Lethal with PARP Inhibitors through the Induction of Homologous Recombination Deficiency. Cancer Cell. 2018, 33, 401–416. [Google Scholar] [CrossRef]
  460. Bhatt, A.P.; Pellock, S.J.; Biernat, K.A.; Walton, W.G.; Wallace, B.D.; Creekmore, B.C.; et al. Targeted inhibition of gut bacterial β-glucuronidase activity enhances anticancer drug efficacy. Proc Natl Acad Sci. 2020, 117, 7374–81. [Google Scholar] [CrossRef]
  461. Hillege, L.E.; Stevens, M.A.M.; Kristen, P.A.J.; Vos-Geelen Jde Penders, J.; Redinbo, M.R.; et al. The role of gut microbial β-glucuronidases in carcinogenesis and cancer treatment: a scoping review. J Cancer Res Clin Oncol. 2024, 150, 495. [Google Scholar] [CrossRef] [PubMed]
  462. Zhao, Z.; Deng, Y.; Han, J.; Ma, L.; Zhu, Y.; Zhang, H.; et al. CircMALAT1 promotes cancer stem-like properties and chemoresistance via regulating Musashi-2/c-Myc axis in esophageal squamous cell carcinoma. MedComm. 2024, 5, e612. [Google Scholar] [CrossRef]
  463. Serna, N.; Álamo, P.; Ramesh, P.; Vinokurova, D.; Sánchez-García, L.; Unzueta, U.; et al. Nanostructured toxins for the selective destruction of drug-resistant human CXCR4+ colorectal cancer stem cells. J Control Release. 2020, 320, 96–104. [Google Scholar] [CrossRef]
  464. Obenauf, A.C.; Zou, Y.; Ji, A.L.; Vanharanta, S.; Shu, W.; Shi, H.; et al. Therapy-induced tumour secretomes promote resistance and tumour progression. Nature. 2015, 520, 368–72. [Google Scholar] [CrossRef]
  465. Wu, P.; Gao, W.; Su, M.; Nice, E.C.; Zhang, W.; Lin, J.; et al. Adaptive Mechanisms of Tumor Therapy Resistance Driven by Tumor Microenvironment. Front Cell Dev Biol. 2021, 9, 641469. [Google Scholar] [CrossRef]
  466. Bayle, A.; Belcaid, L.; Palmieri, L.J.; Teysonneau, D.; Cousin, S.; Spalato-Ceruso, M.; et al. Circulating tumor DNA landscape and prognostic impact of acquired resistance to targeted therapies in cancer patients: a national center for precision medicine (PRISM) study. Mol Cancer. 2023, 22, 176. [Google Scholar] [CrossRef]
  467. Ahronian, L.G.; Corcoran, R.B. Strategies for monitoring and combating resistance to combination kinase inhibitors for cancer therapy. Genome Med. 2017, 9, 37. [Google Scholar] [CrossRef]
  468. Krepler, C.; Xiao, M.; Sproesser, K.; Brafford, P.A.; Shannan, B.; Beqiri, M.; et al. Personalized Preclinical Trials in BRAF Inhibitor–Resistant Patient-Derived Xenograft Models Identify Second-Line Combination Therapies. Clin Cancer Res. 2016, 22, 1592–602. [Google Scholar] [CrossRef] [PubMed]
  469. Pazarentzos, E.; Bivona, T.G. Adaptive stress signaling in targeted cancer therapy resistance. Oncogene. 2015, 34, 5599–606. [Google Scholar] [CrossRef]
  470. Li, L.; Hu, M.; Wang, T.; Chen, H.; Xu, L. Repositioning Aspirin to Treat Lung and Breast Cancers and Overcome Acquired Resistance to Targeted Therapy. Front Oncol. 2020, 9, 1503. [Google Scholar] [CrossRef] [PubMed]
  471. Boe, R.H.; Triandafillou, C.G.; Lazcano, R.; Wargo, J.A.; Raj, A. Spatial transcriptomics reveals influence of microenvironment on intrinsic fates in melanoma therapy resistance. bioRxiv. 6014. [Google Scholar]
  472. Wang, K.; Zhang, X.; Li, A.; Qiao, X.; Xu, Y. The mechanism of action and therapeutic potential of tumor-associated macrophages in tumor immune evasion. Front Immunol. 2025, 16, 1545928. [Google Scholar] [CrossRef]
  473. Kaur, P.; Mohamed, N.E.; Archer, M.; Figueiro, M.G.; Kyprianou, N. Impact of Circadian Rhythms on the Development and Clinical Management of Genitourinary Cancers. Front Oncol. 2022, 12, 759153. [Google Scholar] [CrossRef] [PubMed]
  474. Zhou, I.; Plana, D.; Palmer, A.C. Tumor-specific activity of precision medicines in the NCI-MATCH trial. medRxiv. 2328. [Google Scholar]
  475. Wang, J.; Wang, B.; Chu, H.; Yao, Y. Intrinsic resistance to EGFR tyrosine kinase inhibitors in advanced non-small-cell lung cancer with activating EGFR mutations. OncoTargets Ther. 2016, 9, 3711–26. [Google Scholar] [CrossRef]
  476. Lopez, J.S.; Banerji, U. Combine and conquer: challenges for targeted therapy combinations in early phase trials. Nat Rev Clin Oncol. 2016, 14, 57–66. [Google Scholar] [CrossRef]
  477. Zou, Y.; Zheng, S.; Xie, X.; Ye, F.; Hu, X.; Tian, Z.; et al. N6-methyladenosine regulated FGFR4 attenuates ferroptotic cell death in recalcitrant HER2-positive breast cancer. Nat Commun. 2022, 13, 2672. [Google Scholar] [CrossRef]
  478. Hautaniemi, S.; Kozłowska, E.; Färkkilä, A.; Vallius, T.; Carpén, O.; Kemppainen, J.; et al. Mathematical modeling predicts response to chemotherapy and drug combinations in ovarian cancer. Cancer Res. 2018, 78, canres. [Google Scholar]
  479. Rolfo, C.; Mack, P.C.; Scagliotti, G.V.; Baas, P.; Barlesi, F.; Bivona, T.G.; et al. Liquid Biopsy for Advanced Non-Small Cell Lung Cancer (NSCLC): A Statement Paper from the IASLC. J Thorac Oncol. 2018, 13, 1248–68. [Google Scholar] [CrossRef] [PubMed]
  480. Schneider, L.; Stöckel, D.; Kehl, T.; Gerasch, A.; Ludwig, N.; Leidinger, P.; et al. DrugTargetInspector: An assistance tool for patient treatment stratification. Int J Cancer. 2016, 138, 1765–76. [Google Scholar] [CrossRef] [PubMed]
  481. Vareki, S.M.; Salim, K.Y.; Danter, W.R.; Koropatnick, J. Novel anti-cancer drug COTI-2 synergizes with therapeutic agents and does not induce resistance or exhibit cross-resistance in human cancer cell lines. PLoS ONE. 2018, 13, e0191766. [Google Scholar] [CrossRef]
  482. Aldea, M.; Andre, F.; Marabelle, A.; Dogan, S.; Barlesi, F.; Soria, J.C. Overcoming Resistance to Tumor-Targeted and Immune-Targeted Therapies. Cancer Discov. 2021, 11, 874–99. [Google Scholar] [CrossRef]
  483. Ramirez, M.; Rajaram, S.; Steininger, R.J.; Osipchuk, D.; Roth, M.A.; Morinishi, L.S.; et al. Diverse drug-resistance mechanisms can emerge from drug-tolerant cancer persister cells. Nat Commun. 2016, 7, 10690. [Google Scholar] [CrossRef] [PubMed]
  484. Mead, K.H.; Wang, Y.; Cleary, S.; Arem, H.; Pratt-Chapman, M.L. Defining a patient-centered approach to cancer survivorship care: development of the patient centered survivorship care index (PC-SCI). BMC Heal Serv Res. 2021, 21, 1353. [Google Scholar] [CrossRef]
  485. Jayadevappa, R. Patient Centered Care - A Conceptual Model and Review of the State of the Art. Open Heal Serv Polic J. 2011, 4, 15–25. [Google Scholar] [CrossRef]
  486. Greenup, R.A.; Blitzblau, R.C.; Houck, K.L.; Sosa, J.A.; Horton, J.; Peppercorn, J.M.; et al. Cost Implications of an Evidence-Based Approach to Radiation Treatment After Lumpectomy for Early-Stage Breast Cancer. J Oncol Pr. 2017, 13, JOP. [Google Scholar] [CrossRef]
  487. Jayadevappa, R.; Chhatre, S.; Gallo, J.J.; Malkowicz, S.B.; Schwartz, J.S.; Wittink, M.N. Patient-Centered Approach to Develop the Patient’s Preferences for Prostate Cancer Care (PreProCare) Tool. MDM Polic Pr. 2019, 4, 2381468319855375. [Google Scholar] [CrossRef]
  488. Epstein, A.S.; Desai, A.V.; Bernal, C.; Romano, D.; Wan, P.J.; Okpako, M.; et al. Giving Voice to Patient Values Throughout Cancer: A Novel Nurse-Led Intervention. J Pain Symptom Manag. 2019, 58, 72–79. [Google Scholar] [CrossRef] [PubMed]
  489. Tang, T.C.; Man, S.; Xu, P.; Francia, G.; Hashimoto, K.; Emmenegger, U.; et al. Development of a Resistance-like Phenotype to Sorafenib by Human Hepatocellular Carcinoma Cells Is Reversible and Can Be Delayed by Metronomic UFT Chemotherapy. Neoplasia. 2010, 12, 928–40. [Google Scholar] [CrossRef] [PubMed]
  490. Viossat, Y.; Noble, R. The logic of containing tumors. bioRxiv. 9153. [Google Scholar]
  491. Gallaher, J.A.; Enriquez-Navas, P.M.; Luddy, K.A.; Gatenby, R.A.; Anderson, A.R.A. Data from Spatial Heterogeneity and Evolutionary Dynamics Modulate Time to Recurrence in Continuous and Adaptive Cancer Therapies. 2023. [Google Scholar]
  492. Daley, B.; Kortum, R. Abstract B007: Proximal RTK signaling regulates tumor initiating cell survival and therapeutic responsiveness in EGFR- and KRAS-mutated lung adenocarcinoma. Mol Cancer Res.
  493. Rodriguez, M.J.; Perrone, M.C.; Riggio, M.; Palafox, M.; Salinas, V.; Elia, A.; et al. Targeting mTOR to overcome resistance to hormone and CDK4/6 inhibitors in ER-positive breast cancer models. . 2022. [CrossRef] [PubMed]
  494. Attalla, K.; Sfakianos, J.P.; Galsky, M.D. Genitourinary Cancers. Cancer Treat Res. 2018, 175, 241–58. [Google Scholar]
  495. Maiorano, B.A.; Schinzari, G.; Ciardiello, D.; Rodriquenz, M.G.; Cisternino, A.; Tortora, G.; et al. Cancer Vaccines for Genitourinary Tumors: Recent Progresses and Future Possibilities. Vaccines. 2021, 9, 623. [Google Scholar] [CrossRef]
  496. Mathur, D.; Taylor, B.P.; Chatila, W.; Schultz, N.; Razavi, P.; Xavier, J. Abstract PO-114: Mathematical modeling of tumor heterogeneity to optimize treatment scheduling and delay the evolution of resistance. Cancer Res.
  497. Suresh, S.; Raghavendran, S.; Selvaraj, S. Combining Evolution and Cancer Therapy: A Review of the Mathematical Approach. Curr Cancer Ther Rev. 2022, 18, 7–13. [Google Scholar]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2026 MDPI (Basel, Switzerland) unless otherwise stated